Puneet Kumar, Pran Kishore Deb - Frontiers in Pharmacology of Neurotransmitters-Springer Singapore - Springer (2020)
Puneet Kumar, Pran Kishore Deb - Frontiers in Pharmacology of Neurotransmitters-Springer Singapore - Springer (2020)
Kumar
Pran Kishore Deb Editors
Frontiers in
Pharmacology
of Neurotransmitters
Frontiers in Pharmacology
of Neurotransmitters
Puneet Kumar • Pran Kishore Deb
Editors
Frontiers in Pharmacology
of Neurotransmitters
Editors
Puneet Kumar Pran Kishore Deb
Department of Pharmacology Department of Pharmaceutical Sciences
Central University of Punjab Faculty of Pharmacy
Bathinda, Punjab, India Philadelphia University
Amman, Jordan
Department of Pharmaceutical
Sciences and Technology
Maharaja Ranjit Singh
Punjab Technical University
Bathinda, Punjab, India
This Springer imprint is published by the registered company Springer Nature Singapore Pte Ltd.
The registered company address is: 152 Beach Road, #21-01/04 Gateway East, Singapore 189721,
Singapore
“This book is dedicated to our mentors,
students, and researchers.”
Preface
vii
viii Preface
motivate the young scientists and researchers from the multidisciplinary interna-
tional research community to design innovative drug discovery strategies for the
development of novel therapeutic interventions to cure various neurological
disorders and improve the human health in future.
We thank all the authors for their sincere efforts and valuable contributions in this
book. Dr. Puneet Kumar would like to convey his sincere thanks to Prof. MPS Ishar
(Vice Chancellor, MRSPTU, Bathinda), Prof. S.K. Kulkarni (Emeritus Professor,
Panjab University), and Prof. Anil Kumar (UIPS, Panjab University) for their
valuable guidance and encouragement. Dr. Pran Kishore Deb would also like to
express his gratitude towards his mentor Prof. Raghuprasad M. (Sri Vishnu College
of Pharmacy, AP, India) for his valuable guidance, encouragement, and
contributions; and Dr. Wafa Hourani, Research Assistant Ms. Sara Nidal Abed
(Faculty of Pharmacy, Philadelphia University) for their valuable contributions
during the proofreading of various chapters of the book.
ix
Contents
xi
xii Contents
xv
xvi Editors and Contributors
Contributors
Abstract
Keywords
Neurotransmission · Neurotransmitter · Neuroreceptors · Agonists · Antagonists
P. Kumar (*)
Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical
University, Bathinda, Punjab, India
S. N. Abed · Y. A. Bataineh · M. S. Salem
Faculty of Pharmacy, Philadelphia University, Amman, Jordan
Abbreviations
1.1 Introduction
Neurotransmitters
Glutamate Substance P CO
Epinephrine Serotonin
Aspartate
Norepinephrine Histamine
Dopamine Taurine
The discovery of various types of neurotransmitters has taken place over the past
years where the neurotransmitters have been classified based on their chemical
(Fig. 1.1), functional (Fig. 1.2), and molecular properties along with their location
in the body (Fig. 1.3) (Zhou and Danbolt 2014). Neurotransmitters are classified into
(a) biogenic amines that include serotonin, dopamine, epinephrine (adrenaline), and
norepinephrine (noradrenaline), (b) neuropeptides that include substance P, as well
as (c) amino acids that include glutamate and γ-aminobutyric acid (GABA) (Rangel-
gomez and Meeter 2016). Neurotransmitters show their action via two classes of
receptors possessing distinctive modalities of synaptic transmission. The first class is
known as ionotropic receptors, which comprise the ligand-gated ion channels that
are responsible for eliciting fast synaptic transmission. The second class is
metabotropic receptors, which consist of GPCRs that bind to neurotransmitters
causing slow synaptic transmission by intracellular signaling pathways in addition
to inducing gene expression (Komatsu 2015). The aforementioned neurotransmitters
4 P. Kumar et al.
along with other important ones are briefly discussed in this chapter, taking into
account the roles they serve in the physiological/pathological conditions, including
their mechanisms of action and their pharmacological properties (Fig. 1.4).
1 Neurotransmitters and Their Receptors—State of the Art 5
1.2 GABA
The γ-aminobutyric acid (GABA) is a non-protein amino acid and serves as a key
inhibitory neurotransmitter in the adult brain. GABA is widespread in early embry-
onic life and has been shown to have an excitatory synaptic transmission activity in
the immature brain (Luján et al. 2005). The neurotransmission served by GABA is
known to be present in nearly all organisms, starting from bacteria to human beings
(Owens and Kriegstein 2002). GABA is synthesized by the GABAergic neurons
from L-glutamate, and the catalysis of this reaction is done by the glutamic acid
decarboxylase (GAD) enzyme (Olsen 2002; Bouche et al. 2003). Once GABA is
synthesized, it is then stored in GABAergic synaptic vesicles which is facilitated by
a vesicular GABA transporter (VGAT) and upon depolarization, it undergoes
calcium ion-dependent release. The release of GABA from the presynaptic terminals
will be then followed by the action on GABA receptors (GABAA and GABAB),
(Markwardt and Overstreet-wadiche 2008; Li et al. 2012). GABAA receptors are ion
channels, whereas GABAB receptors are G-protein-coupled receptors (GPCR).
Stimulating GABAA receptor was shown to increase the permeability to chloride
ions which therefore resulted in inhibition or hyperpolarization of neurons by an
increase in the potential of the postsynaptic membrane. Stimulating GABAB recep-
tor leads to modulating cyclic adenosine monophosphate (cAMP) production as well
as increasing the conductance of potassium and hence resulting in either hyperpo-
larization or inhibition of the voltage-gated calcium channels (Olsen 2002; Bouche
et al. 2003).
GABAergic neurotransmission has been shown to play a vital role in
proliferating, migrating, and integrating other neuronal progenitors that are of key
importance for modulating the brain patterns, and deliberates various trophic
functions along with its contribution in the synaptic plasticity (Schmidt and Mirnics
2015). Disruptions in the GABAergic neurotransmission mostly result in sequential
events in the brain development, which was revealed in numerous in vitro and in vivo
studies by using various receptor modulators and knockout (KO) mice models
(Fig. 1.5) (Owens and Kriegstein 2002).
1.3 Glutamate
Glutamine
Pre synaptic terminal
Glutaminase
Mitochondrion
Glutamate
CAD 65/67
GABA
Glutamine
VMAT transporter Astrocyte
Post synaptic receptor and
ionic channel GABA Glutamine
Exocytosis Glutamine
synthetase
GABA Glutamate
Synaptic
cleft
Post synaptic receptor GABA-transporter
and ionic neuron
GABA
GABA-T
Extra synaptic
Post synaptic neuron
Phasic inhibition cleft
Tonic inhibition
(e.g. neuronal excitability)
1.4 Dopamine
Dopamine is considered as the major neurotransmitter in the brain from the cate-
cholamine group. Dopamine is responsible for various functions in the brain includ-
ing controlling the voluntary actions, reward, consciousness, circadian rhythm as
well as cognition (Hasbi et al. 2011). The dopaminergic neurons are present in the
midbrain and the associated regions that include the basal ganglia, ventral tegmental
area, and the retrorubral field (Haber 2016). The mediation of dopamine physiologi-
cal actions is done by its interaction with its receptors over dopaminergic synapses.
There are mainly five types of dopamine receptors that are categorized under two
subclasses, D1 and D2 dopamine receptors (Beaulieu et al. 2015). Dopamine
receptors are typically stable, however sharp and sometimes prolonged. The increase
or decrease in dopamine levels can downregulate or upregulate the number of
dopamine receptors as shown in Fig. 1.7.
8 P. Kumar et al.
1.5 Melatonin
(Wassall et al. 2009). Under stressful conditions, adrenaline is released into the
blood and therefore transmits signals to the organs of the body and creates a specific
response (Daubner et al. 2011). Adrenaline is synthesized from dopamine by the
action of phenylethanolamine N-methyltransferase as shown in Fig. 1.9.
The effects of both noradrenaline and adrenaline were shown to be mediated by
seven transmembrane G-protein-coupled receptors “adrenergic receptors” or
“adrenoceptors” (AR) which can be categorized into two major groups: α (α1, α2)
and β (β1, β2, β3) receptors (Strosberg 1993). The α1-ARs are activated by adrenergic
agonists stimulating Gq protein and phospholipase C (PLC). This activation
promotes the phosphatidylinositol bisphosphate (PIP2) hydrolysis, thereby produc-
ing inositol trisphosphate (IP3) and diacylglycerol (DAG) (Cotecchia 2010). The α2-
AR is referred to as the inhibitory receptors which, upon activation, stimulates the Gi
protein, thus inhibiting adenylate cyclase (AC) and reducing cAMP (Civantos
Calzada and Aleixandre De Artiñano 2001). The β1-AR is responsible for mediating
the cardiovascular responses to the circulating adrenaline as well as noradrenaline
that is released from the sympathetic nerve terminals. The β2-AR is present in the
airway smooth muscle cells including the bronchial muscles. The β3-AR is primarily
present in the adipocytes and can bind to Gi as well as Gs proteins. When these
receptors are stimulated, protein kinase A (PKA) will be activated, L-type Ca++
channels will be phosphorylated, and Ca++ entry will be in the state of relaxation
(Lefkowitz 2000; Ma and Huang 2002; Kohout and Lefkowitz 2003). AR ligands
12 P. Kumar et al.
have been tested and applied as drug therapeutics for the treatment of different
cardiovascular diseases including hypertension, angina pectoris, and congestive
heart failure. AR ligands have been also shown to be useful in the treatment of
diseases like asthma, depression, benign prostatic hypertrophy, glaucoma, shock,
premature labor, opioid withdrawal, and adjunct medications in general anesthesia
(Wassall et al. 2009). Agonists of β2-AR are used as first-line therapeutic agents for
the treatment of asthma and chronic obstructive pulmonary disease (COPD) as
shown in Table 1.1.
1.7 Acetylcholine
Acetylcholine (Ach) is a natural substance and the first neurotransmitter which was
identified by Otto Loewi. Ach alters many functions in the brain like neuronal
excitability, influences transmission, and coordinates the firing of neurons. Ach
also plays an important role in processing memory and learning behavior (Thorne
2010). In Alzheimer’s disease (AD), there is a decrease in the concentration as well
as the function of acetylcholine. Loss of Ach contributes to memory and attention
deficit. It is the major neurotransmitter in the parasympathetic nervous system
(PNS). It acts as a neurotransmitter at various sites, i.e., autonomic ganglia, neuro-
muscular junction, CNS, blood vessels, and postsynaptic receptors in the PNS (Oddo
and Laferla 2006). Acetylcholine is synthesized in cholinergic nerve terminal from
choline. Choline is actively taken up by the axonal membrane by a Na+: choline
transporter. Ach is hydrolyzed by acetylcholinesterase (AchE) (true cholinesterase)
and butyrylcholinesterase (BuchE) (pseudocholinesterase) into choline and acetate.
Acetylcholine acts on cholinergic receptors (muscarinic and nicotinic receptors).
Muscarinic receptors are of five types—M1, M2, M3, M4, M5; and nicotinic
receptors are of two types—Nm and Nn. All muscarinic receptors are G-protein
coupled receptors. The odd-numbered muscarinic receptors act through IP3/DAG
1 Neurotransmitters and Their Receptors—State of the Art 13
1.8 Histamine
Histidine
L-histidine
decarboxy
Histamine
Diamine oxidase
N-methyl transferase
Imidazole acid
N-methyl imidazole
acetic acid riboside
2009; Singh and Jadhav 2013). The H1 receptor is responsible for mediating cellular
migration, nociception, vasodilatation, and bronchoconstriction (Bakker et al. 2001).
On the other hand, the H2 receptor is responsible for modifying the secretion of
gastric acid, production of airway mucus, and vascular permeability (Seifert et al.
2013). The H3 receptor was reported to have a role in neuro-inflammatory diseases
1 Neurotransmitters and Their Receptors—State of the Art 15
(Singh and Jadhav 2013), whereas the H4 receptor has been shown to have a role in
allergy and inflammation (Tiligada 2012; Thurmond 2015).
H1-antihistamines like azatadine, cetirizine, as well as mizolastine are useful for
treating mast cell activated diseases (Church and Church 2013). Other agents like
cimetidine, ranitidine, famotidine, and nizatidine are known as H2R selective
antihistamines that cause a reduction of gastric acid secretion (Shim and Kim
2017). H3R antihistamines include thioperamide, clobenpropit, BF2649,
PF-03654746, JNJ-17216498, and MK 0249. It has been evidenced that histamine
binding to H4 receptors cause exacerbation of allergy and inflammation. It has been
also demonstrated that mast cells have H4 receptors which, upon stimulation,
enhance degranulation and cytokine production. Consequently, antihistamines that
target both the H1 and H4 receptors might be effective treatments for mast cell-
mediated allergic reactions (Mishra et al. 2011).
1.9 Serotonin
Serotonin is a chemical that is found in almost all types of human tissues as well as in
plant and aerobic organisms like bacteria. Serotonin is a ubiquitous monoamine that
acts as both a neurotransmitter and hormone (Mohammad-Zadeh et al. 2008), which
is also known as 5-hydroxytryptamine (5-HT) (Shad 2017). The cell bodies of
serotonergic neuron are localized in the brainstem midline with broad axonal ridge
which extends to all the regions of the CNS (Peterlin and Rapoport 2007). 5-HT is
involved in various diseases like schizophrenia, anxiety, depression, hypertension,
migraine, carcinoid diarrhea, vomiting, irritable bowel syndrome (IBS), pulmonary
hypertension, eating disorders, and others (Pauwels 2003; De Ponti 2004).
5-HT is secreted by the nuclei in the median raphe of the CNS, and then
transferred to the spinal cord and other parts of the brain including the hypothalamus.
The synthesis of 5-HT is initiated through an active uptake of tryptophan in neurons
and enterochromaffin cell by a specific amino acid transporter (Upadhyay 2003;
16 P. Kumar et al.
1.10 Adenosine
Fig. 1.12 Synthesis, biotransformation, and signal transduction mechanism of adenosine and A1,
A2A, A2B, and A3 receptors (reprinted with permission from Pran Kishore Deb 2019a)
et al. 2011; Borea et al. 2018). The primary signal transduction mechanism of ARs
involves the modulation of adenylyl cyclase (AC). Activation of A1 and A3 ARs
causes inhibition of AC activity leading to the reduction of cAMP and inhibition of
protein kinase A (PKA), whereas A2A and A2B ARs activation stimulates the AC
activity and consequently increases cAMP and PKA levels (Fredholm 2014; Merighi
et al. 2018). Figure 1.12 represents the synthesis, biotransformation, and signal
transduction mechanism of adenosine and its four receptor subtypes (Deb 2019a).
Ubiquitous distributions of ARs in the form of homomers, heteromers, or
oligomers have made them interesting drug targets for the therapeutic interventions
of various pathological conditions (Borea et al. 2018; Fredholm 2014; Merighi et al.
2018). In the last two decades a large number of agonists, antagonists, and allosteric
modulators of ARs have been discovered, some of which are under various phases of
clinical trial investigations, including those already in the market as shown in
Fig. 1.13 (Deb 2019a, b, c; Deb et al. 2019a, 2019b, 2018, 2011; Chandrasekaran
et al. 2018; Chandrasekaran et al. 2019; Shaik et al. 2019; Mailavaram et al. 2019;
Al-Attraqchi et al. 2019; Borah et al. 2019; Baraldi et al. 2008). In particular, recent
FDA approval of istradefylline as an A2A AR antagonist for the treatment of
Parkinson’s disease further boosted the research on ARs (Hoffman 2019; Voelker
2019).
18 P. Kumar et al.
NH2 O
O O
N N H O
N N N N
N N
O N N O N N
O
HO
HO HO
Name: Adenosine Name: Theophylline Name: Doxofylline
1. MoA: A1 AR agonist MoA: A1 AR antagonist MoA: A1 AR antagonist
Application: Paroxysmal supraventricular Application: Asthma Application: Asthma
tachycardia (PSVT)
2. MoA: A2A AR agonist
Application: Myocardial perfusion imaging
NH2
OH O
N N
N N N
O NH N
N N
O N N O
N O N O
N HO
N O
O N
HO
HO
Fig. 1.13 Therapeutic applications of clinically approved drugs targeting ARs (reprinted with
permission from Pran Kishore Deb 2019a)
1.11 Angiotensin
Angiotensinogen
Cathepsin D Renin
Angiotensin I
Chymase ACE
Angiotensin II
AT 1
Aminopeptides A
AT 2
Angiotensin III
Nitric oxide (NO) production results from the oxidation of the terminal guanidine
nitrogen of the amino acid arginine. The catalysis of this reaction occurs by the
action of NADPH-dependent enzyme, nitric oxide synthase (NOS). When NO is
synthesized, it gets diffused outside the cell. It is produced by a group of enzymes
called nitric oxide synthases. These enzymes convert arginine into citrulline, pro-
ducing NO in the process as shown in Fig. 1.15 (Virarkar et al. 2013).
The concentration of NO differs under physiological conditions (Tieu et al.
2003). As a neurotransmitter, NO was shown to play a role in the activation of the
cGMP-dependent protein kinase G (PKG) pathway which is responsible for
phosphorylating synaptophysin, which is of significant importance for the fusion
of presynaptic vesicles, therefore leading to the potentiation and facilitating of
neurotransmission (Wang et al. 2005). NO was also shown to play a role in the
inhibition of GABAergic synaptic transmission by cGMP-dependent pathways in
addition to ion channels and exchangers (Yamamoto et al. 2015). NO plays a role in
supporting vascular homeostasis in endothelium-dependent vasodilatation; however,
its over- or underproduction was shown to be related to pathological conditions
(Džoljić et al. 2015). NO is therefore considered to be a critical mediator under
certain pathological conditions. For example, in brain ischemia-reperfusion injury,
1 Neurotransmitters and Their Receptors—State of the Art 21
1.13 Opioids
used for pain control. Well-known opioid antagonists are nalorphine, naloxone, and
naltrexone. Opioids like morphine and others produce their activity by the release of
the EOPs through direct action on opioid receptors (Koneru et al. 2009).
1.14 Cannabinoids
1.15 Conclusion
References
Ahluwalia J et al (2000) Cannabinoid 1 receptors are expressed in nociceptive primary sensory
neurons. Neuroscience 100(4):685–688
Al-Attraqchi O, Attimarad M, Venugopala K, Nair A, Al-Attraqchi N (2019) Adenosine A2A
receptor as a potential drug target-current status and future perspectives. Curr Pharm Des
25:2716–2740
Appendino G, Chianese G (2011) Cannabinoids: occurrence and medicinal chemistry. Curr Med
Chem 18(7):1085–1099
Axelrod J (1974) The pineal gland: a neurochemical transducer. Science 184(4144):1341–1348
Baker KM, Booz GW, Dostal DE (1992) Cardiac actions of angiotensin II: role of an intracardiac
renin-angiotensin system. Annu Rev Physiol 54(1):227–241
Bakker RA et al (2001) Histamine H(1)-receptor activation of nuclear factor-kappa B: roles for G
beta gamma- and G alpha(q/11)-subunits in constitutive and agonist-mediated signaling. Mol
Pharmacol 60(5):1133–1142
Baraldi PG, Tabrizi MA, Gessi S, Borea PA (2008) Adenosine receptor antagonists: translating
medicinal chemistry and pharmacology into clinical utility. Chem Rev 108:238–263
Bauer JH, Reams GP (1986) Renal effects of angiotensin converting enzyme inhibitors in hyper-
tension. Am J Med 81(4):19–27
Beaulieu J, Espinoza S, Gainetdinov RR (2015) Dopamine receptors–IUPHAR review 13. Br J
Pharmacol. Wiley Online Library 172(1):1–23
Berger M, Gray JA, Roth BL (2009) The expanded biology of serotonin. Annu Rev Med
60:355–366. https://doi.org/10.1146/annurev.med.60.042307.110802
Biegański T (1983) Biochemical, physiological and pathophysiological aspects of intestinal
diamine oxidase. Acta Physiol Pol 34(1):139–154
Bochkov DV et al (2012) Shikimic acid: review of its analytical, isolation, and purification
techniques from plant and microbial sources. J Chem Biol 5:5–17. https://doi.org/10.1007/
s12154-011-0064-8
Boonstra TW et al (2007) Effects of sleep deprivation on neural functioning: an integrative review.
Cell Mol Life Sci 64(7–8):934–946. https://doi.org/10.1007/s00018-007-6457-8
Borah P, Deka S, Mailavaram RP, Deb PK (2019) P1 receptor agonists/antagonists in clinical trials-
potential drug candidates of the future. Curr Pharm Des 25:2792–2807
24 P. Kumar et al.
Borea PA, Gessi S, Merighi S, Varani K (2016) Adenosine as a multi-signalling guardian angel in
human diseases: when, where and how does it exert its protective effects? Trends Pharmacol Sci
37:419–434
Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K (2018) Pharmacology of adenosine receptors:
the state of the art. Physiol Rev 98:1591–1625
Borg L, Kreek MJ (2003) The pharmacology of opioids. In: Graham A et al (eds) Principles of
addiction medicine. Chevy Chase, MD, American Society of Addiction Medicine, pp 141–155
Bouche N, Lacombe B, Fromm H (2003) GABA signaling: a conserved and ubiquitous mechanism.
Trends Cell Biol 13(12):607–610
Chandrasekaran B, Deb PK, Kachler S, Akkinepalli RR, Mailavaram R, Klotz K-N (2018)
Synthesis and adenosine receptors binding studies of new fluorinated analogues of pyrido
[2,3-d] pyrimidines and quinazolines. Med Chem Res 27:756–767
Chandrasekaran B, Samarneh S, Jaber AMY, Kassab G, Agrawal N (2019) Therapeutic potentials
of A2B adenosine receptor ligands: current status and perspectives. Curr Pharm Des
25:2741–2771
Chen J-F, Eltzschig HK, Fredholm BB (2013) Adenosine receptors as drug targets—what are the
challenges? Nat Rev Drug Discov 12:265
Church MK, Church DS (2013) Pharmacology of antihistamines. Indian J Dermatol 58(3):219–224
Civantos Calzada B, Aleixandre De Artiñano A (2001) Alpha-adrenoceptor subtypes. Pharmacol
Res. https://doi.org/10.1006/phrs.2001.0857
Cogan MG (1990) Angiotensin II: a powerful controller of sodium transport in the early proximal
tubule. Hypertension 15(5):451–458
Cotecchia S (2010) The α1-adrenergic receptors: diversity of signaling networks and regulation. J
Recept Sig Transduct Res 30(6):410–419. Taylor & Francis
Dahlitz M et al (1991) Delayed sleep phase syndrome response to melatonin. Lancet 337
(8750):1121–1124
Danbolt NC, Furness DN, Zhou Y (2016) Neuronal vs glial glutamate uptake: resolving the
conundrum. Neurochem Int 98:29–45. Elsevier Ltd
Daubner SC, Le T, Wang S (2011) Tyrosine hydroxylase and regulation of dopamine synthesis.
Arch Biochem Biophys 508(1):1–12. NIH Public Access
De Ponti F (2004) Pharmacology of serotonin: what a clinician should know. Gut 53:1520–1535
Deb PK (2019a) Therapeutic potentials of adenosine receptors: the state of the art. Curr Pharm Des
25:2789–2791
Deb PK (2019b) Progress in the development of agonists, antagonists and allosteric modulators of
adenosine receptors. Curr Pharm Des 25:2695–2696
Deb PK (2019c) Recent updates in the computer aided drug design strategies for the discovery of
agonists and antagonists of adenosine receptors. Curr Pharm Des 25:747–749
Deb PK, Balakumar C, Rao AR, Roy PP, Roy K (2011) QSAR of adenosine receptor antagonists:
exploring physicochemical requirements for binding of pyrazolo[4,3-e]-1,2,4-triazolo[1,5-c]
pyrimidine derivatives with human adenosine A3 receptor subtype. Bioorg Med Chem Lett
21:818–823
Deb PK, Mailavaram R, Chandrasekaran B, Kaki VR, Kaur R, Kachler S, Karl-Norbert K,
Akkinepally RR (2018) Synthesis, adenosine receptor binding and molecular modeling studies
of novel thieno[2,3-d]pyrimidine derivatives. Chem Biol Drug Des 91:962–969
Deb PK, Chandrasekaran B, Mailavaram R, Tekade RK, Muttaleb A, Jaber Y (2019a) Molecular
modeling approaches for the discovery of adenosine A2B receptor antagonists : current status
and future perspectives. Drug Discov Today 24:1854–1864
Deb PK, Deka S, Borah P, Abed SN, Klotz K (2019b) Medicinal chemistry and therapeutic
potential of agonists, antagonists and allosteric modulators of A1 adenosine receptor: current
status and perspectives. Curr Pharm Des 25:2697–2715
Deussen A (2000) Metabolic flux rates of adenosine in the heart. Naunyn Schmiedeberg's Arch
Pharmacol 362:351–363
1 Neurotransmitters and Their Receptors—State of the Art 25
Jin X et al (2003) Targeted disruption of the mouse Mel1b melatonin receptor. Mol Cell Biol 23
(3):1054–1060. https://doi.org/10.1128/MCB.23.3.1054
Jinwala FN, Gupta M (2012) Synthetic cannabis and respiratory depression. J Child Adolesc
Psychopharmacol 22(6):459–462. https://doi.org/10.1089/cap.2011.0122
Jou M-J, Peng T-I (2018) Protective stabilization of mitochondrial permeability transition and
mitochondrial oxidation during mitochondrial Ca2+ stress by melatonin’s cascade metabolites
C3-OHM and AFMK in RBA1 astrocytes. J Pineal Res:e12538. https://doi.org/10.1111/jpi.
12538
Jou M-J et al (2007) Melatonin protects against common deletion of mitochondrial
DNA-augmented mitochondrial oxidative stress and apoptosis. J Pineal Res 43:389–403.
https://doi.org/10.1111/j.1600-079X.2007.00490.x
Jutel M, Akdis CA (2007) Histamine as an immune modulator in chronic inflammatory responses.
Clin Exp Allergy 37:308–310
Kagami S et al (1994) Angiotensin II stimulates extracellular matrix protein synthesis through
induction of transforming growth factor-beta expression in rat glomerular mesangial cells. J Clin
Invest 93(6):2431–2437
Karasek M, Winczyk K (2006) Melatonin in humans. J Physiol Pharmacol 57:19–39
Kavalali ET (2015) The mechanisms and functions of spontaneous neurotransmitter release. Nat
Rev Neurosci 16(1):5–16. Nature Publishing Group
Kohout TA, Lefkowitz RJ (2003) Regulation of G protein-coupled receptor kinases and arrestins
during receptor desensitization. Mol Pharmacol 63(1):9–18
Komatsu H (2015) Novel therapeutic GPCRs for psychiatric disorders. Int J Mol Sci:14109–14121.
https://doi.org/10.3390/ijms160614109
Koneru A, Satyanarayana S, Rizwan S (2009) Endogenous opioids: their physiological role and
receptors. Global J Pharmacol 3(3):149–153
Lefkowitz RJ (2000) The superfamily of heptahelical receptors. Nat Cell Biol 2(7):E133–E136
Lerner AB et al (1958) Isolation of melatonin, the pineal gland factor that lightens melanocytes. J
Am Chem Soc 58:2587
Leurs R et al (2009) Molecular and biochemical pharmacology of the histamine H4 receptor. Br J
Pharmacol 157:14–23
Li Y et al (2012) A novel role of intestine epithelial GABAergic signaling in regulating intestinal
fluid secretion. Am J Physiol Gastrointest Liver Physiol 303(4):453–460. https://doi.org/10.
1152/ajpgi.00497.2011
Liu J et al (2016) MT1 and MT2 melatonin receptors: a therapeutic perspective. Annu Rev
Pharmacol Toxicol 56:361–383
Liu J et al (2019) (Pro) renin receptor regulates lung development via the Wnt/B-catenin signaling
pathway. Am J Physiol Lung Cell Mol Physiol 317(2):202–211
Lochner A, Marais E, Huisamen B (2018) Melatonin and cardioprotection against ischaemia/
reperfusion injury: what’s new? A review. J Pineal Res 65(1):e12490
Luján R, Shigemoto R, López-Bendito G (2005) Review glutamate And GABA receptor signalling
in the developing brain. Neuroscience 130(3):567–580
Lv C et al (2018) Dopamine D2-like receptors (DRD2 and DRD4) in chickens: Tissue distribution,
functional analysis, and their involvement in dopamine inhibition of pituitary prolactin expres-
sion. Gene 651:33–43. Elsevier
Ma YC, Huang XY (2002) Novel signaling pathway through the beta-adrenergic receptor. Trends
Cardiovasc Med 12(1):46–49
Mackie K (2006) Cannabinoid receptors as therapeutic targets. Annu Rev Pharmacol Toxicol
46:101–122. https://doi.org/10.1146/annurev.pharmtox.46.120604.141254
Mailavaram R, Al-Attraqchi O, Kar S, Ghosh S (2019) Current status in the design and develop-
ment of agonists and antagonists of adenosine A3 receptor as potential therapeutic agents. Curr
Pharm Des 25:2772–2787
Majidinia M et al (2018) The role of melatonin, a multitasking molecule, in retarding the processes
of ageing. Ageing Res Rev 47:198–213. https://doi.org/10.1016/j.arr.2018.07.010
1 Neurotransmitters and Their Receptors—State of the Art 27
Manchester LC et al (2000) High levels of melatonin in the seeds of edible plants Possible function
in germ tissue protection. Life Sci 67:3023–3029
Manchester LC et al (2015) Melatonin: an ancient molecule that makes oxygen metabolically
tolerable. J Pineal Res 59:403–419. https://doi.org/10.1111/jpi.12267
Markwardt S, Overstreet-wadiche L (2008) GABAergic signalling to adult-generated neurons. J
Physiol 586(16):3745–3749. https://doi.org/10.1113/jphysiol.2008.155713
Merighi S, Gessi S, Borea PA (2018) Adenosine receptors: structure, distribution, and signal
transduction. In: Borea P, Varani K, Gessi S, Merighi S, Vincenzi F (eds) The adenosine
receptors, The receptors, vol 34, pp 33–57
Mishra GP et al (2011) Recent patents and emerging therapeutics in the treatment of allergic
conjunctivitis. Recent Patents Inflamm Allergy Drug Discov 5:26–36
Mohammad-Zadeh LF, Moses L, Gwaltney-Brant SM (2008) Serotonin: a review. J Vet Pharmacol
Ther 31:187–199
Morgan PJ et al (1994) Melatonin receptors: localization, molecular pharmacology and physiologi-
cal significance. Neurochem Int 24(2):101–146
Murrough JW, Abdallah CG, Mathew SJ (2017) Targeting glutamate signalling in depression:
progress and prospects. Nat Rev Drug Discov 16(7):472. Nature Publishing Group
Nabavi SM et al (2019) Anti-inflammatory effects of melatonin: a mechanistic review. Crit Rev
Food Sci Nutr 59(sup1):4–16. https://doi.org/10.1080/10408398.2018.1487927
Naganuma F et al (2017) Histamine N-methyltransferase regulates aggression and the sleep-wake
cycle. Sci Rep 7(1):15899. https://doi.org/10.1038/s41598-017-16019-8
Nathan C (1997) Inducible nitric oxide synthase: what difference does it make? J Clin Invest
100:2417–2423
Ni W, Watts SW (2006) 5-hydroxytryptamine in the cardiovascular system: focus on the serotonin
transporter (SERT). Clin Exp Pharmacol Physiol 33:575–583
Nyberg L et al (2016) Dopamine D2 receptor availability is linked to hippocampal–caudate
functional connectivity and episodic memory. Proc Natl Acad Sci 113(28):7918–7923. National
Acad Sciences
O’Mahony L, Akdis M, Akdis CA (2013) Regulation of the immune response and inflammation by
histamine and histamine receptors. J Allergy Clin Immunol 128(6):1153–1162
Oddo S, Laferla FM (2006) The role of nicotinic acetylcholine receptors in Alzheimer’s disease. J
Physiol Paris 99(2–3):172–179
Olsen R (2002) GABA’. In: Charney D et al (eds) Neuropsychopharmacology the fifth generation
of progress. Cambridge University Press, Cambridge, pp 159–168
Onaolapo AY, Onaolapo OJ (2018) Circadian dysrhythmia-linked diabetes mellitus: examining
melatonin’s roles in prophylaxis and management. World J Diabetes 9(7):99–114
Owens DF, Kriegstein AR (2002) Is there more to GABA than synaptic inhibition? Nat Rev
Neurosci 3:715–727
Pacheco R, Lejeune M, Climent N, Oliva H, Gatell JM, Gallart T, Mallol J (2005) CD26, adenosine
deaminase, and adenosine receptors mediate costimulatory signals in the immunological syn-
apse. PNAS 102:9583–9588
Pauwels PJ (2003) 5-HT receptors and their ligands. Neuropharmacology 1083:38–50
Peterlin BL, Rapoport AM (2007) Clinical pharmacology of the serotonin receptor agonist,
zolmitriptan. Expert Opin Drug Metab Toxicol 3(6):899–912
Poeggeler B (2005) Melatonin, aging, and age-related diseases. Endocrine 27(2):201–212
Pytliak M et al (2011) Serotonin receptors – from molecular biology to clinical applications. Physiol
Res 60:15–25
Rangel-Barajas C, Coronel I, Florán B (2015) Dopamine receptors and neurodegeneration. Aging
Dis 6(5):349. JKL International LLC
Rangel-gomez M, Meeter M (2016) Neurotransmitters and novelty: a systematic review. J
Psychopharmacol 30(1):1–13. https://doi.org/10.1177/0269881115612238
Rasakham K (2008) Kappa Opioid Receptor regulation of ERK1/2 MAP kinase signaling cascade:
molecular mechanisms modulating cocaine reward. Northeastern University, Boston, MA
28 P. Kumar et al.
Thorne BM (2010) Neurotransmitters’. In: Weiner IB, Craighead WE (eds) The corsini encyclope-
dia of psychology. Wiley, Hoboken, NJ, pp 1–2
Thurmond RL (2015) The histamine H4 receptor: from orphan to the clinic. Front Pharmacol 6
(65):1–11
Tieu K, Ischiropoulos H, Przedborski S (2003) Nitric oxide and reactive oxygen species in
Parkinson’s disease. IUBMB Life 55(6):329–335. https://doi.org/10.1080/
1521654032000114320
Tiligada E (2012) Editorial: Is histamine the missing link in chronic inflammation. J Leukoc Biol
92:4–6
Tordjman S et al (2017) Melatonin: pharmacology, functions and therapeutic benefits. Curr
Neuropharmacol 15(3):434–443
Upadhyay SN (2003) Serotonin receptors, agonists and antagonists. IJNM 18(1 & 2):1–11
Virarkar M et al (2013) L-arginine and nitric oxide in CNS function and neurodegenerative
diseases. Crit Rev Food Sci Nutr 53(11):1157–1167. https://doi.org/10.1080/10408398.2011.
573885
Voelker R (2019) Add-on drug approved for “Off” episodes of Parkinson disease. JAMA
322:1246–1246
Von Gall C, Stehle JH, Weaver DR (2002) Mammalian melatonin receptors: molecular biology and
signal transduction. Cell Tissue Res 309:151–162. https://doi.org/10.1007/s00441-002-0581-4
Walker MC, van der Donk WA (2016) The many roles of glutamate in metabolism. J Ind Microbiol
Biotechnol 43(2–3):419–430. Springer
Wang H et al (2005) Presynaptic and postsynaptic roles of NO, cGK, and RhoA in long-lasting
potentiation and aggregation of synaptic proteins. Neuron 45(3):389–403. https://doi.org/10.
1016/j.neuron.2005.01.011
Wassall RD, Teramoto N, Cunnane TC (2009) Noradrenaline. In: Encyclopedia of neuroscience.
Academic Press, Cambridge, pp 1221–1230
Wu H et al (2019) Beyond a neurotransmitter: The role of serotonin in inflammation and immunity.
Pharmacol Res 140:100–114. https://doi.org/10.1016/j.phrs.2018.06.015
Yaksh TL, Wallace MS (2011) Opioids, analgesia, and pain management. In: Goodman and
Gilman’s the pharmacological basis of therapeutics. McGraw-Hill Medical, New York, NY,
pp 481–526
Yamamoto K et al (2015) Presynaptic cell type-dependent regulation of GABAergic synaptic
transmission by nitric oxide in rat insular cortex. Neuroscience 284:65–77. IBRO
Yoshikawa T et al (2013) molecular mechanism of histamine clearance by primary human
astrocytes. Glia 61:905–916. https://doi.org/10.1002/glia.22484
Zhdanova IV, Lynch HJ, Wurtman RJ (1997) Melatonin: a sleep-promoting hormone. Sleep 20
(10):899–907
Zhou Y, Danbolt NC (2014) Glutamate as a neurotransmitter in the healthy brain. J Neural Transm
121(8):799–817. https://doi.org/10.1007/s00702-014-1180-8
Zimmermann H (2000) Extracellular metabolism of ATP and other nucleotides. Naunyn
Schmiedeberg's Arch Pharmacol 362:299–309
Zisapel N (2001) Circadian rhythm sleep disorders pathophysiology and potential approaches to
management. CNS Drugs 15(4):311–328
Drug-Receptor Interactions
2
Balakumar Chandrasekaran, Haneen Al-Joubi, Sara Samarneh,
Ghadir Kassab, Pran Kishore Deb, Puneet Kumar, Bilal A. Al-Jaidi,
Yazan Al-Thaher, and Yazan A. Bataineh
Abstract
chemical aspects involved between the drug and the receptor. Further, we have
shed light on the development of adenosine receptor antagonists through in silico
interactions as a case study.
Keywords
Abbreviations
3D Three dimensional
Ach Acetylcholine
ARs Adenosine receptors
ATP Adenosine triphosphate
CRC Concentration-response curve
DBD DNA-binding domain
DRC Dose-response curve
GPCRs G-protein-coupled receptors
H-bond Hydrogen bonding
LBD Ligand-binding domain
PPARγ Peroxisome proliferator-activated receptor gamma
RTK Receptor tyrosine kinase
TK Tyrosine kinase
vdW Van der Waals
2.1 Introduction
Upon drug administration, it gets absorbed, transported to the site of action through
general circulation, and elicits a pharmacological response. The target site for the
drug action is ultimately a bio-macromolecule, known as a receptor. Receptors are
mostly membrane-bound proteins, consist of various amino acids, and receive
signals from outside the cell through a ligand molecule. When such small molecules
or ligands bind to the receptor and interact, subsequently they elicit some form of
cellular/tissue responses (Uings and Farrow 2000). There are three different ways
wherein the receptor responds to the chemical messenger/ligand such as relay of
signal, amplification, and integration. Most of the receptors are integral proteins
connected to the protein-lipid bilayer of the cell membrane. The two functionally
important components of receptors include the recognition component (ability of
recognizing specific molecules) and the amplification component (an ability of the
intermolecular complex formation between the ligand and the receptor) to initiate a
pharmacological response (Pierce et al. 2002). Based on the complementarity
between the ligand and binding site of a receptor, each receptor is connected to a
specific biochemical pathway by specific binding towards a particular ligand. Upon
ligand binding to its binding site present in the receptor, it may activate or inhibit
various receptor-associated biochemical events. Further, this single receptor will
2 Drug-Receptor Interactions 33
Unused binding
DRUG region DRUG Extra
interaction
Drug extension
Receptor Receptor
Fig. 2.1 The binding of a drug to the binding site of the receptor based on shape complementarity
The structure of receptors varies which depends entirely on the diversity of the
binding site and functions of the receptors. Principally, four types of receptors are
identified, namely ligand-gated ion channel receptors, guanine nucleotide binding
regulatory protein (G-protein)-coupled receptors (GPCRs), tyrosine kinase-linked
receptors, and nuclear receptors.
Fig. 2.2 The signaling process shown by ligand-gated ion channel receptors
GPCRs are the largest family of receptors for some transmitters (metabotropic
glutamate and dopamine), light-sensitive compounds, various hormones, lipids,
glycoproteins, small molecules, peptides, and larger proteins. Upon binding of
these ligands to the GPCRs, they facilitate the interaction with members of the
G-protein receptor family. GPCRs are mostly identified in eukaryotes, yeasts, and
animals (Saengsawang and Rasenick 2015). The structure of GPCRs includes seven
transmembrane α-helices, and loops for their integration. As the name indicates,
these receptors are coupled to diversified intracellular-effector systems incorporating
both extracellular (a binding site for large ligand) and intracellular (separate binding
site) involving the activation of signal transduction pathways resulting in a cellular
response, accordingly (Rosenbaum et al. 2009). Most of the modern medicines
(about 40%) act through modulating the signaling processes associated with
GPCRs (Chandrasekaran et al. 2019). Since they occur as transmembrane proteins,
they are difficult to isolate, purify, and crystallize. To date, bovine rhodopsin,
β-adrenoceptors, and A2A adenosine receptors were crystallized in their pure forms
(Deb et al. 2019a). A typical orientation of a type II receptor involved in the signal
transduction process is shown in Fig. 2.3.
NH3+
GPCR
Cell membrane
COO-
α-helix. The TK receptor is located on the surface of the cell exhibiting higher
affinity for many of the ligands such as growth factors, hormones, and cytokines
(Cadena and Gill 1992). They are the key regulators for a number of cellular
biochemical processes including their critical role in the progression of cancer.
There are almost twenty different classes of TK-linked receptors and the insulin
receptor is one such example. The main function of TK-linked receptors is the
phosphorylation of tyrosine amino acid residue of target proteins by transferring a
phosphate group from high energy donor molecule (ATP) (Huang and Reichardt
2003). The receptor tyrosine kinase (RTK) pathway is regulated through a number of
positive and negative feedback loops. Since RTKs are involved in many cellular
functions (cell proliferation, differentiation, survival, metabolism, migration, and
cell cycle regulation), they must be regulated to avert cellular abnormalities like
fibrosis and cancer (Gschwind et al. 2004). Figure 2.4 illustrates a typical signaling
process mediated by kinase-linked receptors.
They are normally present in the cytoplasm when there is no ligand binding to the
receptor. Once the ligand binds to such receptors, they migrate to the nucleus of the
cell, accordingly. They are composed of different binding domains such as
N-terminal regulatory domain, DNA-binding domain (DBD) containing two zinc
fingers, hinge region, ligand-binding domain (LBD), and C-terminal domain. In
particular, the N-terminal domain interacts with other cellular transcription factors in
a ligand-independent manner (Robinson-Rechavi and Laudet 2003). Depending on
such interactions, the binding/activity of the receptor can be affected, suitably.
Nuclear receptors are accountable for the recognition of steroidal and thyroid
hormones, fatty acids, bile acids, prostaglandins, and certain vitamins. As they
regulate the gene expression by binding directly to DNA, they are classified as
transcription factors and all these processes depend on the availability of a ligand.
Upon ligand binding to a nuclear receptor, conformational changes occur in the
receptor which activates the receptor; thereby upregulation or downregulation of
gene expression has been effected. Hence, they control the general embryonic
development, homeostasis, and metabolism (Aranda and Pascual 2001). Lipophilic
36
Receptor tyrosine
kinase
cytokine
RAF-1 Cytokine
p
p p receptor
MAPKK p p p
p Non receptor
p p p
MAPK p tyrosine
p
kinase
p p
MAP kinase STAT
MAPK signal
pathway
JAK/STAT signal
p p p pathway
STAT
Drug targets are molecules such as proteins (enzymes, receptors, and transport
proteins), lipids, carbohydrates, or nucleic acids (as RNA or DNA) wherein the
drug is going to reach and bind. An important step for a drug action is the binding
and subsequent interaction with the target. While drug is traveling throughout the
body, it reaches its target and identifies the correct binding site in a receptor protein
and interacts to elicit a biological response. The binding site in a receptor also known
as a pocket or canyon is at the surface of the macromolecular target. It is very
important to gain an insight into the forces involved in the receptor-drug binding in
order to determine the mode of drug action (Hase et al. 2009). A receptor has unique
amino acids with specific side chains, so the functional groups in drugs have the
complementary shape characteristics to fit into the binding site or pocket. There are
points for the attachment between a drug and the receptor selectively to provoke the
drug action, and this binding always exists in an equilibrium state. The drug should
have strong interaction with the receptor for association and to allow signal trans-
duction; at the same time, it should also show weak interaction for dissociation and
to allow the drug to depart, after completion of its action. Hence, a fine balance
between strong and weak interactions is required for the better activity of the drugs
(Bs Suvarna 2011).
38 B. Chandrasekaran et al.
where, K on is the rate constant for the drug-receptor complex formation and depends
on the concentrations of both the drug and the receptor and K off is the rate constant
for dissociation or the breakdown of the complex and entirely depends on the
concentration of the drug-receptor complex. The pharmacological response of a
drug is associated with its affinity towards the receptor. Moreover, the stability of
the drug-receptor complex can be determined based on the dissociation of the
complex as represented in Eq. 2.2.
½drug½receptor
Kd ¼ ð2:2Þ
½durg‐receptor complex
After studying the different types of interactions between the drugs and their
receptors, scientists have proposed and developed a set of hypothesis over the
years, known as theories of drug-receptor interactions. These theories can explain
the ability of the drug to interact with the receptor to induce or stimulate a suitable
biological response (Kenakin 2008).
This theory was proposed by Gaddum and Clark in 1926 (Clark 1926). According to
this theory, the drug binds to its specific receptor (complementary structures like
lock and key) and gives a cellular response. Based on the study, scientists concluded
that the intensity of cellular response is directly proportional to the number of
receptors occupied which means that a maximum response occurs when all receptors
are occupied. Drugs exhibit all or no response on each receptor, so it will be either
fully activated or not at all activated. Moreover, there are no partial activation
because once the drug-receptor complex dissociates, the response will halt, accord-
ingly. This concept is illustrated in Fig. 2.6.
As this theory does not cover the concept of partial agonist, Ariens and
Stephenson (Stephenson 1956) have modified the occupancy theory to incorporate
the concept of partial agonist. They have used the main concept of the drug-receptor
interactions that generally involves two stages, namely, affinity and efficacy. Affin-
ity is a feature that determines the capacity of a drug to bind to the receptor for
activating and producing a desired response. The degree of affinity between the drug
and receptor depends on molecular complementarity. If drugs have different affinity
(Dissociation)
No response
A B
100% 100%
Drug 1
Drug 2
% Response
% Response
Drug 3
Drug 1
Drug 2
Drug 3
0% 0%
Fig. 2.7 (A) Drugs have similar affinities but different efficacies; (B) drugs have similar efficacies
but different affinities
to specific receptors, then it is difficult to figure out which one is going to bind to the
receptor. The answer is when a drug with high binding capacity comes around the
receptor, it will inevitably bind to the receptor, even if it is associated with another
drug. In this situation, the former drug will remove another drug which has lower
affinity from the active site of the receptor. The nature of the binding or the
interaction between the receptor and the drug molecule relies mainly on the degree
of affinity (Shuker et al. 1996). On the other hand, the maximum response can be
achieved by the dose of the drug, termed as efficacy and intrinsic activity (Nelson
et al. 2016). Figure 2.7 elucidates the exact difference between the affinity and the
efficacy.
Figure 2.7A illustrates the percentage of biological response relative to the dose
of three different drugs, and all of them have similar affinities to the same receptor.
At the same time, they have quite different efficacies ranging from 100% and ended
with approximately 70% of the maximum. By taking into consideration that the drug
which reached 100% of the maximum response is known as a full agonist while
others are partial agonists. On the other hand, Fig. 2.7B presents the dose-response
curve for three drugs, all of which exhibit an equal efficacy but the affinities of each
one vary. It is also interesting to know that those three drugs are full agonists.
Moreover, both agonist and antagonist will bind to receptors but each one will
produce a different response. Agonist will activate the receptor, while antagonist
will halt the receptor action. Also, a drug may be an agonist when it is bound to one
of the receptors and exerts a positive response, while the same drug could be
antagonist for another receptor. The reason why it is possible for two drugs that
can fit a specific receptor exert different effects is not clarified by this modified
occupancy theory (Maehle et al. 2002). The concept of efficacy has been displayed
in Fig. 2.8.
2 Drug-Receptor Interactions 41
Efficacy
Fig. 2.9 The concept of ligands (agonist, partial agonist, and antagonist)
The rate theory was proposed by the scientist Paton in 1961 (Paton 1961). This
theory suggested that the pharmacological activity of any drug depends on the rate of
association and dissociation of the drug with the receptor and will differ based on
agonist, partial agonist, or antagonist. Further, the intensity of the pharmacological
response depends on the number of the drug-receptor interactions per unit time, and
this is directly proportional to the rate of association and dissociation between the
drug and the receptor and the total number of molecules involved in such
interactions. Like in the occupancy theory, the rate theory was also unable to justify
why various types of compounds display their own features. The concept of agonist,
partial agonist, and antagonist has been presented in Fig. 2.9.
42 B. Chandrasekaran et al.
Based on the consideration of the flexibility of the receptor, two general types of
macromolecular perturbation were proposed by the scientist Belleau (Belleau 1964).
Either one or both of them could result into the receptor functions once the interac-
tion between the drug and the receptor has occurred. According to the macromolec-
ular perturbation theory, the intensity of pharmacological response is directly
proportional to the rate of formation of perturbations. One type of perturbation is a
specific conformational perturbation which contributes to generate a biological
response when specific molecules bind to the receptor’s binding site (agonist). The
other type is nonspecific conformational perturbations. In this case, no response will
be produced because the receptor binds with other types of molecule (antagonist).
2 Drug-Receptor Interactions 43
However, if the molecule has both molecular perturbations, the result will be a
mixture of two complexes (partial agonist). This theory illustrates the physicochem-
ical properties of the molecules binding to the receptor, but does not cover the
significance of the concept of inverse agonism.
This theory has been developed because the activation-aggregation theory men-
tioned previously did not sufficiently explain the activation of a receptor. Hence, the
two-state model of receptor activation theory was developed which depends on the
competitive and noncompetitive kinetics. In addition, it is based on the performance
and results of experiments applied by direct binding to the receptor’s binding-site. In
this model, scientists relied on the previously proposed models but explained the
process of activation in a comprehensive way (Bridges and Lindsley 2008). Simply,
the theory assumes the presence of the receptor in two conformational shapes. The
first one makes the receptor in the active state (R) and then can give a downstream
effect. While the second concept keeps the receptor in an inactive (R) state; therefore
no pharmacological effects can be exerted. The receptor will be in the equilibrium
state if the ligand is not available in the binding site. This is a state between R
44 B. Chandrasekaran et al.
and R, which is known as the basal activity of the receptor. Depending on the
constant need for the formation of a drug-receptor complex (Kd and Kd), the drug
will either bind to one conformational state or to both of them (Colquhoun 1998).
Ligands such as full agonist, partial agonist, full inverse agonist, and antagonist will
bind with a specific state and give a completely different effect. In the following
subsections, the behavior of different ligands is discussed briefly.
Agonist
Giving maximum
effect
Active receptor
Fig. 2.10 The agonist binding selectively to the receptor in the active state
2 Drug-Receptor Interactions 45
receptor and shift the equilibrium to inactive state, thus decreasing the basal activity
of the receptor and a negative efficacy was observed (<0% efficacy).
Antagonist
Inactive receptor
The driving force for drug-receptor interaction is the low energy state of the drug-
receptor complex. The biological activity is related to the drug affinity for the
receptor, i.e., the stability of the complex. Dissociation constant of the drug-receptor
complex gives an idea about how potent is the drug. The binding interactions occur
through points of attachment; for a chemical compound they are the functional
groups. Functional groups use their electronic and shape characters in the binding
process. If we talk about reversible binding, binding of the drug to receptor should be
in equilibrium state. Drug-target interactions can be grouped into two types: Perma-
nent (irreversible)—covalent bonding and reversible interactions. All plausible
interactions existing between a receptor and the ligand are discussed in the following
subsections.
A covalent bond is produced between two species by mutual sharing of electrons and
is the strongest bond, irreversible, and exhibits a stability of 40 to 110 kcal/mol.
This type of covalent bond is mostly observed in drug-enzyme or drug-DNA
complexes rather than the drug-receptor complex. It is also beneficial in avoiding
toxic effects of drugs through an irreversible inhibition of the receptor. This covalent
bond usually occurs between a nucleophile (molecule having negative charge, rich in
electrons) such as hydroxyl or thiol group in the receptor amino acids and an
electrophile (molecule having positive charge, deficient in electrons) such as epoxide
or allyl group in drug structure (Kumalo et al. 2015). Two types of covalent bonds,
namely polar and nonpolar, are possible during interactions. Water molecule is an
example of a polar covalent bond, whereas peptide bond is an example of
noncovalent bond. The antibiotic penicillin is an irreversible inhibitor of the enzyme
glycopeptide-transpeptidase, the enzyme which catalyzes an essential step in bacte-
rial cell wall synthesis. Penicillin covalently blocks the active-site amino acid serine
present inside the glycopeptide-transpeptidase through the formation of a
covalent bond.
Ionic bond is also known as electrostatic interaction which is weaker than the
covalent bond and can provide an ionic interaction energy of 5 kcal/mol. This
bond appears between two opposite (negative and positive) charges of amino acids
of a receptor and ionized species of the ligand (Klebe 2013). Under the normal
physiological pH, some of the basic amino acids like arginine, lysine, and histidine
bearing amino group in their side chain get protonated, thereby providing a cationic
environment. On the other hand, acidic amino acids such as aspartic acid and
glutamic acid having carboxylic group get deprotonated and become anionic in
2 Drug-Receptor Interactions 47
nature. Thus, both positive (cationic) and negative (anionic) charges available in the
receptors take part in an ionic bonding with ionized groups of drugs. Ionic bond is a
most prevalent bond in drug-receptor interaction which depends entirely on the
extent of ionization and the distance between two opposite charges. An example
of ionic interaction is presented in Fig. 2.12.
Due to the electronegativity of hetero-atoms (O, N, S, and halogens over the carbon
atom), an electric dipole is formed subsequently generating the polarization in
bonds. In the polarized bond, one of the pole will be partially positive and the
other confers partially negative charge, respectively. These partially positive or
negative charges can form an electrostatic bond with either partially charged atoms
or ionized elements. As a result of higher electronegativity of one atom, an asym-
metric distribution of the electrons was observed. Hence, ion-dipole interaction
involves an ion (side chain amino acids of receptors) and a dipole (drug) or vice
versa, but dipole-dipole interaction occurs between two dipoles of the drug and
receptor, respectively. This bond is polar and electrostatic; also the dipole-dipole
interaction is weaker than ion-dipole interaction. This type of interaction involves
the energy of 1 to 7 kcal/mol (Du et al. 2016). An example for the drug is
zaleplon (Fig. 2.13) which is indicated for the treatment of insomnia.
N N
Ion-dipole interaction
N
O
O O
2.90 Å O
O H
and a hydroxyl group proton (H-O) is 2.75 Å, whereas a carbonyl oxygen (C¼O)
and proton of N-H is 2.90 Å (Fig. 2.14).
A hydrogen bond is very important and unique to hydrogen atom, exclusively as
it is the only atom that can confer a positive charge at physiological pH (Balakumar
et al. 2010). Hydrogen (H)-bonding includes two types of interactions, that is,
intermolecular and intramolecular H-bonds, respectively. However, compounds
that tend to make intramolecular H-bond will be less active and unable to interact
with the receptor. The possibility of hydrogen bonding involves the orientation of
hydrogen atom in donor and acceptor group and depends on the distance between
two atoms (1.5–2.2 Å). Hydrogen bond acceptor has electron-rich atom and slightly
negative (carboxylate ion) charge, whereas the donor has an electro-deficient hydro-
gen and slightly positive charge (alkyl ammonium ion or secondary and primary
amines) (Kuhn et al. 2010). Two types of hydrogen bonding interactions are
presented in Fig. 2.15.
Charge transfer interactions happen between electron donor group in one molecule
(alkene) and an electron acceptor in another group (aromatic ring). Some amino
acids in the receptor have electron donor groups like -OH group in aromatic amino
acid tyrosine and carboxylate group (-COO) of aspartate (Zhang et al. 2016).
Similarly, few amino acids bearing electron acceptor group (sulfur-containing
amino acid cysteine) and amino acid residues like histidine, tryptophan, and
2 Drug-Receptor Interactions 49
O
H
Intramolecular H-bonding
O
O Receptor
H O
H
Intermolecular H-bonding
CN
Cl Cl
Cl Cl
HO
CN
Fig. 2.16 Chlorthalonil interacting with tyrosine residue of the receptor through charge transfer
process
Hydrophobic interaction
Water/solvent
Nonpolar chain of
receptor Receptor
Nonpolar
nonpolar Drug nonpolar chain
chain in
Drug chain
Π-Π interaction
Fig. 2.18 Aromatic system of lacosamide drug interacting with the aromatic ring of the receptor
through π-π interaction
Lipid bilayer
Protein
Fig. 2.19 Van der Waals interaction between the drug and the receptor is shown for epinephrine
including H-bonding and ionic bonding
Hydrophobic
Charge Transfer N
Hydrogen bond
H Ionic, ion-dipole
N
O N
Hydrophobic Hydrophobic
Hydrophobic
O
Dipole-dipole, Hydrophobic
H-bond,
or Halogen bond
Dibucaine
Fig. 2.20 Multiple interactions shown by Dibucaine towards the receptor
2.6.1 Agonists
Some endogenous molecules are responsible for the regulation of certain physiolog-
ical functions through binding with receptors and interact in the tissues which lead to
a specific physiological response. Like a natural messenger to the receptor, an
agonist is a chemical messenger that can activate the receptor as a result of binding
to it. Due to the structural similarity with the natural messenger, agonists can also
exhibit intermolecular interactions/bonds by employing the same induced fit similar
to natural messenger performs. However, the binding of agonist to the receptor
should be a reversible binding. The knowledge about the properties of the binding
site including the geometry and topography, the chemical structure of the normal
substrate that binds to the receptor, the correct binding group of agonists, position of
interaction with complementary binding region, and the shape and size of agonists is
required for the design of novel agonists so as to fit the binding site of the receptor
(Auerbach 2016).
There are two terms which describe the agonists as partial and full based on dose-
response curve (DRC) or concentration-response curve (CRC). This curve is
obtained initially by administering endogenous compounds like Ach to animal
muscle tissue and allowing the contraction of muscle as a measure of response.
Initially, the concentration of Ach is low; therefore, only a small number of
molecules interact with the receptor, and at the time they had linear relationship
between the concentration and the response because all of the receptors are occupied
at 100%. If a similar kind of response is obtained by the use of an exogenous
compound to this muscle tissue, then such compound is known as a full agonist.
However, 50% response indicates that the administered compound is a partial
52 B. Chandrasekaran et al.
Full agonist
in presence of
partial agonist
Effect
50%
Partial agonist
0%
Agonist concentration
agonist (Lambert 2004). The CRC pattern of different types of agonists is shown in
Fig. 2.21.
2.6.2 Antagonists
Receptor antagonists are compounds that can inactivate the receptor after binding to
it. The antagonist must also have the complementary shape and binding group to
orient towards the binding site of a receptor. In general, the chemical structure of
antagonists exhibits slightly higher size than the endogenous compound and is
classified into competitive and noncompetitive antagonists. If the administration of
Ach does not initiate a response in the presence of another exogenous compound or
there is a need for a higher concentration of Ach, it indicates that the exogenous
compound could be a competitive antagonist. It directly demonstrates that the
agonist and the antagonist compete each other for the same binding site. The
competitive antagonist possesses structural similarity to agonists in terms of size,
shape, and functional groups to allow binding on the same agonist binding site on the
receptor. As mentioned earlier, the response will be tardy after adding an increased
amount of the agonist; consuming the binding is reversible. The predominant
binding to a receptor depends on the higher concentration of either agonist or an
antagonist (Buchwald 2017). If the unknown drug exhibits 50% of response and no
further enhanced response obtained even with the addition of excess amount of Ach,
it is a noncompetitive antagonist. Figure 2.22 represents the CRC of both competi-
tive and noncompetitive antagonists.
There are two strategies to design the noncompetitive antagonists such as alloste-
ric antagonists (Fig. 2.23) and antagonists using umbrella effect (Fig. 2.24). If the
designed compound binds to an allosteric binding site (another site of the normal
agonist binding site) that may be located beside the major binding site, then it leads
to geometric changes in the binding site, thereby preventing the agonist binding is
called as noncompetitive antagonist. It could be a drug or an endogenous compound,
2 Drug-Receptor Interactions 53
A B
100% Agonist alone
100% Agonist alone
% Response
% Response
Agonist + antagonist
50%
50%
Agonist + antagonist
Conformational
Protein change Protein
Allosteric
site
Regulatory
Regulatory molecule
molecule
antagonist displays a part of its structure like a tail that will cover the opening of the
binding site, thus preventing the binding of an agonist (Karschin et al. 1988). In this
case, the percentage of antagonism depends on the amount of both the agonist and
the antagonist.
The strength of the association between the receptor and a drug can be determined by
total free energy of interaction. Nevertheless, it does not clarify about the quality of
interaction or the effect of the addition of new functional group. Hence, it is very
important to estimate the contribution of an individual functional group to drug-
receptor interactions. To understand drug-protein interactions, functional group
additives (Eq. 2.3) or the additivity of free enthalpy components (Eq. 2.4) is majorly
employed (Andrews et al. 1984).
The free energy of binding is defined in terms of the binding energies for the
individual functional groups that construct a drug molecule according to Eq. 2.5.
ΔG ¼ TΔSt,r þ nr Er ¼ E Nx Ex . . . . . . ð2:5Þ
where TΔSt,r is the loss of overall translational and rotational entropy related to the
drug binding, nr is the number of internal degrees of conformational freedom lost on
binding the drug molecule, and Er is the energy equivalent of the entropy loss
associated with the loss of each degree of conformational freedom on receptor
binding (Andrews et al. 1984).
If the specific functional group of a drug aligned to the specified functional group of
the receptor without any strain, the Ex is known as intrinsic binding energy or
apparent binding energy. In general, Ex is the combination of the various enthalpic
and entropic interactions including enthalpy of interaction between the drug and
receptor binding site. The change in enthalpy is associated with the removal of water
of hydration (the functional group and its target binding site), subsequent bond
formation between the displaced water molecules, and the corresponding entropy
terms associated with the displacement and subsequent bonding of water molecules.
Thus, intrinsic binding potentials can be used reasonably in an additive manner in the
determination of the drug-receptor interaction (Jencks 1981).
2 Drug-Receptor Interactions 55
Based on Eq. 2.5, the binding energy Ex can be determined by comparing the
binding energies for pairs of compounds that differ only in terms of functional
group “X.” This concept was employed by the scientist “Page” who declared it as
“Anchor Principle” (Page 1977). It is based on the fact that the difference in binding
of a drug molecule by the presence or the absence of the particular functional group
is mainly due to the number of factors associated with that functional group. In other
words, the binding energy Ex with loss of any degrees of conformational freedom
arose due to the binding of group “X.” The magnitude of the binding energies
deduced by the anchor principle will vary according to the quality of the interaction.
If the functional groups are unable to align correctly, then small or even repulsive
interaction may occur.
The scientist Page (1977) determined that the intrinsic binding energies for the CH2
group (methylene) fall in the range of 12–14 kJ/mol based on the data of the
selectivity of amino acid-tRNA synthetases. Under physiological conditions, the
value of Gibb’s free energy is approximated (in kJ/mol) by Eq. 2.6.
For example, the calculated binding energies for isoleucine and its desmethyl
analog (Fig. 2.25) to isoleucyl-tRNA synthetase are 29.7 and 15.9 kJ/mol,
respectively, demonstrating that the methyl group contributes a total of 13.8 kJ/mol
to overall interactions. In case of long chain hydrocarbons, the positive contribution
is observed mainly because of dispersion forces and hydrophobic interactions
generating the loss of conformational entropy on binding. Hence, 3 kJ/mol binding
energy average derived for sp2 and sp3 carbons is same to the “average” decrease in
free energy of binding. While comparing with the unsaturated or cyclic analogs, this
effect will be higher in case of saturated hydrocarbons (Andrews et al. 1984).
Some acidic and basic entities of charged groups influence the binding interaction
between the ligand and its receptor. In particular, the phosphate (cation) binds to
OH
H H
HN N HN N
N N N N
O O
HO OH HO OH
HO HO
They are the type of stereoisomers, but not mirror images of each other.
Diastereomers are actually the complexes formed between two enantiomers, thereby
yielding different energies and chemical properties consequently resulting in differ-
ent dissociation constants for drug-receptor complexes of enantiomeric drugs. There
are a special case of diastereomers such as geometric isomers (E and Z) and epimers
(compounds having the same chemical formula but differing in their spatial
arrangements around the single carbon atom). They can be separated conveniently
using chromatography or recrystallization techniques than the enantiomers.
Diastereomers exhibit different energies and stabilities due to the fact that they
demonstrate different interactions with the same receptor after binding to the binding
site (Kier 1997). For example, the neuroleptic potency of the Z-isomer of the
chlorprothixene (an antipsychotic drug) is 12 times greater than that of the
corresponding E-isomer. Conversely, the E-isomer of the diethylstilbestrol
(an anticancer drug) had 14 times better estrogenic activity than the corresponding
Z-isomer (Fig. 2.27).
58 B. Chandrasekaran et al.
S S
(Z)-Chlorprothixene (E)-Chlorprothixene
highly active less active
HO
HO
OH
HO
(E)-Diethyl stilbesterol (Z)-Diethyl stilbesterol
highly active less active
According to the nomenclature of Ariëns (1987), the potent isomer is called as the
eutomer and the weaker one is known as distomer. The potency ratio of higher
affinity enantiomer to lower affinity is termed as eudismic ratio. The distomer can be
regarded as an impurity in the mixture, which may contribute to undesirable side
effects or toxicity. In some cases, the distomer will be responsible for the biological
activity and the eutomer attributable to the side effects. D-ketamine (Fig. 2.28), a
hypnotic and analgesic agent, is responsible for the pharmacological actions,
whereas the isomer L-ketamine is known for the undesired side effects. It is also
possible that both isomers are active biologically, but one of them causes toxicity
(e.g., the local anesthetic prilocaine). In some cases, it is required to have the two
isomers for better pharmacological activity. Both isomers of bupivacaine (Fig. 2.28)
act as local anesthetic, but only the L-isomer shows vasoconstrictive activity (Aps
and Reynolds 1978). On the other hand, the D-isomer is responsible (i.e., eutomer)
for both the diuretic activity and the side effect (uric acid retention).
Enantiomers may have different therapeutic actions, for example, Darvon, an
analgesic drug and its enantiomer, Novrad is an antitussive drug. Thus, these
enantiomers are marketed (Darvon and Novrad) separately under different trade
names. Another case for enantiomers is that they may display opposite effects. The
(R) enantiomer of 1-methyl-5-phenyl-5-propylbarbituric acid (Fig. 2.28) acts as a
narcotic, while the (S) enantiomer works as a convulsant. Hence, the receptor has an
ability to select and recognize the isomers through the chiral nature. Enantiomers
may have different biological activities depending on the fact that one isomer may fit
into the receptor binding site much better than its counterpart to demonstrate better
2 Drug-Receptor Interactions 59
Cl Cl
NH NH
O O O NH
(S)-Ketamine (R)-Ketamine N
more active-hypnotic more active-hypnotic
(RS)-Bupivacaine
local anesthetic
O O
N N
O N O O N O
H H
(R)-1-methyl-5-phenyl-5- (S)-1-methyl-5-phenyl-5-
propylbarbituric acid propylbarbituric acid
narcotic analgesic convulsant
pharmacological activity profile (Arthur 1927). If a receptor has two binding site
points (Fig. 2.29 A, B), then it is difficult to recognize the specific enantiomer
(epinephrine); on the other hand, if a receptor has three binding site points
(Fig. 2.29 C, D), it can recognize a particular enantiomer and distinguishes between
pairs of enantiomers (Arthur 1927).
R-()-isomer of epinephrine has three points of interaction due to the specific
conformation to maximize molecular complementarity. However, the S-(+)-isomer
showed two sites of interaction (the hydroxyl group cannot interact with the binding
site) and exhibited lower binding energy (Fig. 2.29A, B).
Similarly, Talapatra et al. studied the crystal structure of the Eg5-K858 complex
and its implications in structure-based design of thiadiazole-containing inhibitors as
anticancer agents (Talapatra et al. 2018). In their study, the inhibitor molecule K-858
(Fig. 2.30) exists in a racemic mixture, which was resolved using chiral HPLC.
Interestingly, S-enantiomer of K-858 showed higher inhibition of Eg5 protein, while
R-enantiomer was unable to inhibit the protein. Figure 2.30 describes the favorable
interaction for the S-enantiomer of K-858 displaying the correct orientation of
methyl group to solvent accessible region, while the phenyl ring is involved in
aromatic π-π interaction (Trp127) and hydrophobic interactions (Arg119 and
Pro137). Thus, the S-enantiomer demonstrated good inhibition of Eg5 protein and
acts as a potential anticancer agent. On the other hand, the phenyl group with a
significantly larger hydrophobic character than the methyl would be placed towards
the solvent region resulting in larger unfavorable interactions for the R-enantiomer
which is responsible for the lack of inhibition of Eg5 protein.
60 B. Chandrasekaran et al.
HO HO
H OH
HO NH2 HO NH2
OH
Ar Ar
R-(-)-Epinephrine S-(+)-Epinephrine
A B
HO HO
H OH
HO NH2 HO NH2
OH
Ar Ar H
H
R-(-)-Epinephrine S-(+)-Epinephrine
C D
Fig. 2.29 Binding of epinephrine enantiomers to two-site receptor (A, B a, b) and three-site
receptor (C, D c, d)
Solvent O Solvent
region region O
N N Unfavorable
O
steric crash N N
S N O
H S N
H
Aromatic Π-Π interaction (Trp127)
Larger unfavorable interaction
Hydrophobic interaction (Arg119 and Pro137)
No activity
Favorable interaction for good activity
S-isomer of K-858 R-isomer of K-858
active Inactive
Fig. 2.30 Binding interactions of enantiomers of K-858 with crucial amino acids in the binding
site of Eg5 protein
2 Drug-Receptor Interactions 61
Conformational isomers are the type of isomers formed due to the free rotation of
single bonds and cannot be separated. The concept of pharmacophore is best
described in terms of the configuration of a set of atoms and the bio-active confor-
mation as well (Balakumar et al. 2018). The crucial amino acid residues in the
binding site of the receptor can bind to only one specific conformer. The conformer
that binds to the receptor’s binding site should have adequate energy which can be
determined by sophisticated instrumentations such as X-ray crystallography and
NMR spectrophotometry or by computation through molecular mechanics
calculations. It is very imperative to identify the bioactive conformation which is
an active conformation of the drug that is involved in binding to the receptor for the
design of ideal drug candidates. If there is a lead compound exhibiting low potency,
then it is mainly due to the existence of a low amount of the active conformer in a
solution. For example, the antidiabetic drug rosiglitazone (Fig. 2.31) binding to
peroxisome proliferator-activated receptor gamma (PPARγ); the favorable orienta-
tion must be in a “U” shaped conformation for better activity (Gampe et al. 2000). In
this particular conformation, the thiazolidinone moiety was buried into the binding
site so that it can display H-bonding interactions with crucial amino acids (Ser289
and Tyr473). Thus, the bioactive conformer of rosiglitazone demonstrated good
inhibition of PPARγ.
Li and Biel demonstrated the correlation between the conformers and the
tranquilizing action of 4-(4-Hydroxypiperidino)-40 -fluorobutyrophenone
(Fig. 2.32A) with mild anti-emetic property (Li and Biel 1969). The compound
had 2 chair (Fig. 2.32B,E ) and 2 twisted-boat (Fig. 2.32C, D) conformations.
Initially, a relative compound, N-methyl-4-piperidinol (R ¼ Me), was considered,
and the difference in free energy between the axial and equatorial hydroxyl
conformers was determined to be 0.94 0.05 kcal/mol at 40 C (the equatorial
conformer is most favorable by a factor of 4.56 over axial conformer). The energies
for the twist-boat conformers are 6 kcal/mol higher due to hydrogen bonding; thus
C was considered as more stable than B. Based on this assumption, three
O O
HN N N
O
S
Rosiglitazone
Fig. 2.31 2D structure of rosiglitazone and 3D interaction plot of rosiglitazone with PPARγ
62 B. Chandrasekaran et al.
OH
O
N
F
A
4-(4-Hydroxypiperidino)-4'-fluorobutyrophenone
B C D E
F G H
conformationally rigid chair analogs (F-H f–h) were synthesized and evaluated for
their muscle relaxant activity. The structure G was found to be conformationally less
stable with the axial hydroxyl group resulting in a better molecular complementarity
with the receptor, thereby yielding good pharmacological activity.
This is a distinguished approach that can be used to determine the bioactive
conformation of a drug molecule in the drug-receptor complex. This involves the
synthesis of conformationally rigid analogs, followed by biological evaluation. The
highest potent analog can be used as a prototype. The major disadvantage in this
approach is that in order to form the analogs, additional atoms must be added to the
original compound and this may affect both chemical and physical properties. In
such cases, it is essential that the drug and the analog must be similar in size, shape,
and mass.
Adenosine receptors (ARs) are classified into four subtypes A1, A2A, A2B, and A3
ARs belonging to the superfamily of GPCRs (Kaur et al. 2011; Agrawal et al. 2019;
Deb 2019a). At present, about 40% of modern medicines function through the
mediation of various signaling processes associated with GPCRs (Deb et al.
2 Drug-Receptor Interactions 63
2.10 Conclusion
Certainly, for the drug to function, it must interact with the target protein such as
receptors. The interactions between a drug and its receptor are mediated through the
structural features, mainly by functional groups. It is very imperative that probing the
real receptors for the establishment of essential binding features can offer crucial
information about the mode of drug action. Of equal importance, the binding
orientation of the drug structure inside the binding site of a receptor can influence
the pharmacological activity. Theories of drug-receptor interactions can provide
more useful information to deduce the probability of proposing the mode of action
of any drug under biological environment. In particular, noncovalent interactions
mostly are weaker and they work in conjunction with other types of interactions.
This is mainly due to the loss in translational entropy (first interaction) followed by
lower entropy (second interaction). Finally, the effect of this cooperativity causes the
conversion of weak interactions to yield strong interactions.
The selectivity of the drug is primarily decided by the strength and existence of
good interactions between the functionalities in drug-receptor complexes. Many
64 B. Chandrasekaran et al.
Fig. 2.33 Ligand interaction pattern showing percentage of contacts with crucial amino acid
residues of A3 AR
times charged functional groups tend to bind more effectively than polar functional
groups, which bind more tightly than nonpolar groups. In case of electrostatic
interactions, ammonium ions are efficient followed by phosphate ion, and then
carboxylate anion. In order to exhibit good pharmacological activity of the ligands,
they must have higher binding energy than the calculated average binding energy.
Conversely, compounds demonstrating less binding energy can fit into receptors
poorly. Hence, the determination of drug-receptor interactions is very essential for
the safety and efficacy of a drug.
Acknowledgments The authors wish to thank Prof. Mutaz Sheikh Salem (President) and Prof.
Marwan Kamal (University Counsellor) of Philadelphia University, Jordan, for the support and
research funding to BC (467/34/100 PU).
References
Agrawal N, Chandrasekaran B, Al-Aboudi A (2019) Recent advances in the in-silico structure-
based and ligand-based approaches for the design and discovery of agonists and antagonists of
A2A adenosine receptor. Curr Pharm Des 25:774–782
Andrews PR, Craik DJ, Martin JL (1984) Functional group contributions to drug-receptor
interactions. J Med Chem 27:1648–1657
2 Drug-Receptor Interactions 65
Aps C, Reynolds F (1978) An intradermal study of the local anaesthetic and vascular effects of the
isomers of bupivacaine. Br J Clin Pharmacol 6:63–68
Aranda A, Pascual A (2001) Nuclear hormone receptors and gene expression. Physiol Rev
81:1269–1304
Ariëns EJ (1987) Stereochemistry in the analysis of drug-action. Part II. Med Res Rev 7:367–387
Arthur RC (1927) Biological relations of optically isomeric substances. Nature 120:152
Auerbach A (2016) Dose-response analysis when there is a correlation between affinity and
efficacy. Mol Pharmacol 89:297–302
Baillie TA (2016) Targeted covalent inhibitors for drug design. Angew Chemie Int Ed
55:13408–13421
Balakumar C, Lamba P, Pran Kishore D et al (2010) Synthesis, anti-inflammatory evaluation and
docking studies of some new fluorinated fused quinazolines. Eur J Med Chem 45:4904–4913
Balakumar C, Narayana BL, Kishore DP et al (2012) Design, microwave-assisted synthesis and in
silico docking studies of new 4H-pyrimido[2,1-b]benzothiazole-2-arylamino-3-cyano-4-ones as
possible adenosine A2B receptor antagonists. Indian J Chem Sect B Org Med Chem Chem
51:1105–1113
Balakumar C, Ramesh M, Tham CL et al (2018) Ligand- and structure-based in silico studies to
identify kinesin spindle protein (KSP) inhibitors as potential anticancer agents. J Biomol Struct
Dyn 36:3687–3704
Banda V, Chandrasekaran B, Köse M et al (2013) Synthesis of novel pyrido[3,2-e]- [1,2,4]triazolo
[1,5-c]pyrimidine derivatives: potent and selective adenosine A3 receptor antagonists. Arch
Pharm (Weinheim) 346:699–707
Barratt E, Bingham RJ, Warner DJ et al (2005) Van der Waals interactions dominate
ligandprotein association in a protein binding site occluded from solvent water. J Am Chem
Soc 127:11827–11834
Belleau B (1964) A molecular theory of drug action based on induced conformational perturbations
of receptors. J Med Chem 7:776–784
Bridges TM, Lindsley CW (2008) G-Protein-coupled receptors: from classical modes of modula-
tion to allosteric mechanisms. ACS Chem Biol 3:530–541
Buchwald P (2017) A three-parameter two-state model of receptor function that incorporates
affinity, efficacy, and signal amplification. Pharmacol Res Perspect 5:e00311
Cadena DL, Gill GN (1992) Receptor tyrosine kinases. FASEB J 6:2332–2337
Chandrasekaran B, Deb PK, Rao R (2017) Structure-based design and pharmacological study of
fluorinated fused quinazolines as adenosine A2B receptor antagonists. JSM Chem 5:1–10
Chandrasekaran B, Deb PK, Kachler S et al (2018) Synthesis and adenosine receptors binding
studies of new fluorinated analogues of pyrido[2,3-d]pyrimidines and quinazolines. Med Chem
Res 27:756–767
Chandrasekaran B, Samarneh S, Jaber AMY, Kassab G, Agrawal N (2019) Therapeutic potentials
of A2B adenosine receptor ligands: current status and perspectives. Curr Pharm Des
25:2741–2771
Clark AJ (1926) The reaction between acetyl choline and muscle cells. J Physiol 61:530–546
Colquhoun D (1998) Binding, gating, affinity and efficacy: the interpretation of structure-activity
relationships for agonists and of the effects of mutating receptors. Br J Pharmacol 125:923–947
Deb PK (2019a) Editorial: Progress in the development of agonists, antagonists and allosteric
modulators of adenosine receptors. Curr Pharm Des 25:2695–2696
Deb PK (2019b) Recent updates in the computer aided drug design strategies for the discovery of
agonists and antagonists of adenosine receptors. Curr Pharm Des 25:747–749
Deb PK (2019c) Therapeutic potentials of adenosine receptors: the state of the art. Curr Pharm Des
25(26):2789–2791
Deb PK, Mailavaram R, Chandrasekaran B et al (2018) Synthesis, adenosine receptor binding and
molecular modelling studies of novel thieno[2,3-d]pyrimidine derivatives. Chem Biol Drug Des
91:962–969
66 B. Chandrasekaran et al.
Deb PK, Chandrasekaran B, Mailavaram R et al (2019a) Molecular modeling approaches for the
discovery of adenosine A2B receptor antagonists: current status and future perspectives. Drug
Discov Today 24:1854–1864
Deb PK, Deka S, Borah P, Abed SN, Klotz K-N (2019b) Medicinal chemistry and therapeutic
potential of agonists, antagonists and allosteric modulators of A1 adenosine receptor: current
status and perspectives. Curr Pharm Des 25:2697–2715
Du X, Li Y, Xia YL et al (2016) Insights into protein–ligand interactions: mechanisms, models, and
methods. Int J Mol Sci 17:144
Gampe RT, Montana VG, Lambert MH et al (2000) Asymmetry in the PPARγ/RXRα crystal
structure reveals the molecular basis of heterodimerization among nuclear receptors. Mol Cell
5:545–555
Gschwind A, Fischer OM, Ullrich A (2004) The discovery of receptor tyrosine kinases: targets for
cancer therapy. Nat Rev Cancer 4:361–370
Hase T, Tanaka H, Suzuki Y et al (2009) Structure of protein interaction networks and their
implications on drug design. PLoS Comput Biol 5:e1000550
Huang EJ, Reichardt LF (2003) Trk receptors: roles in neuronal signal transduction. Annu Rev
Biochem 72:609–642
Jencks WP (1981) On the attribution and additivity of binding energies. Proc Natl Acad Sci U S A
78:4046–4050
Karlin A (1967) On the application of “a plausible model” of allosteric proteins to the receptor for
acetylcholine. J Theor Biol 16:306–320
Karschin A, Aizenman E, Lipton SA (1988) The interaction of agonists and noncompetitive
antagonists at the excitatory amino acid receptors in rat retinal ganglion cells in vitro. J Neurosci
8:2895–2906
Kati WM, Wolfenden R (1989) Contribution of a single hydroxyl group to transition-state discrim-
ination by adenosine deaminase: evidence for an “entropy trap” mechanism. Biochemistry
28:7919–7927
Kaur R, Kishore DP, Narayana BL et al (2011) A facile microwave-assisted synthesis of
8,9-cycloalkathieno[3,2-e] [1,2,4]triazolo[1,5-c]pyrimidin-5(6H )-ones. J Chem Sci 123:69–73
Kenakin T (2008) Receptor theory. Curr Protoc Pharmacol 41:1.2.1–1.2.28
Kier LB (1997) In: Wolff ME (ed) Burgers’ medicinal chemistry and drug discovery, vol 4. Wiley,
New York, NY, pp 5355–5423
Klebe G (2013) Protein–ligand interactions as the basis for drug action. In: Klebe G (ed) Drug
design. Springer, Berlin, Heidelberg, pp 61–88
Koshland DE (1958) Application of a theory of enzyme specificity to protein synthesis. Proc Natl
Acad Sci U S A 44:98–104
Koshland DE, Neet KE (1968) The catalytic and regulatory properties of enzymes. Annu Rev
Biochem 37:359–411
Kuhn B, Mohr P, Stahl M (2010) Intramolecular hydrogen bonding in medicinal chemistry. J Med
Chem 53:2601–2611
Kumalo HM, Bhakat S, Soliman MES (2015) Theory and applications of covalent docking in drug
discovery: merits and pitfalls. Molecules 20:1984–2000
Lambert DG (2004) Drugs and receptors. BJA Educ 4:181–184
Leff P (1995) The two-state model of receptor activation. Trends Pharmacol Sci 16:89–97
Leff P, Scaramellini C, Law C, McKechnie K (1997) A three-state receptor model of agonist action.
Trends Pharmacol Sci 18:355–362
Levine D, Reid TW, Wilson IB (1969) Free energy of hydrolysis of the phosphoryl-enzyme
intermediate in alkaline phosphatase-catalyzed reactions. Biochemistry 8:2374–2380
Lew MJ, Ziogas J (2004) The two-state model of antagonist-AT1 receptor interaction: an hypothesis
defended but not tested. Biochem Pharmacol 67:397–399
Li JP, Biel JH (1969) Steric structure-activity relationship studies on a new butyrophenone deriva-
tive. J Med Chem 12:917–919
2 Drug-Receptor Interactions 67
Maehle A-H, Prüll C-R, Halliwell RF (2002) The emergence of the drug receptor theory. Nat Rev
Drug Discov 1:637–641
Mailavaram RP, Al-Attraqchi OHA, Ghosh SK (2019) Current status in the design and develop-
ment of agonists and antagonists of adenosine A3 receptor as potential therapeutic agents. Curr
Pharm Des 25:2772–2787
Marc J (2008) Pharmacogenetics of drug receptors. EJIFCC 19:48–53
Monod J, Wyman J, Changeux J-P (1965) On the nature of allosteric transitions: a plausible model.
J Mol Biol 12:88–118
Nelson MR, Johnson T, Warren L et al (2016) The genetics of drug efficacy: opportunities and
challenges. Nat Rev Genet 17:197–206
Page MI (1977) Entropy, binding energy, and enzymic catalysis. Angew Chem Int Ed Engl
16:449–459
Paton WDM (1961) A theory of drug action based on the rate of drug-receptor combination. Proc R
Soc London Ser B Biol Sci 154:21–69
Pierce KL, Premont RT, Lefkowitz RJ (2002) Seven-transmembrane receptors. Nat Rev Mol Cell
Biol 3:639–650
Pramanik D (2015) Receptor. Principles of physiology. 3rd edition, Jaypee Brothers Medical
Publishers, India, pp 661-669.
Pran Kishore D, Balakumar C, Raghuram Rao A et al (2011) QSAR of adenosine receptor
antagonists: exploring physicochemical requirements for binding of pyrazolo[4,3-e]-1,2,4-
triazolo[1,5-c]pyrimidine derivatives with human adenosine A3 receptor subtype. Bioorg Med
Chem Lett 21:818–823
Robinson-Rechavi M, Laudet V (2003) Bioinformatics of nuclear receptors. Methods Enzymol
364:95–118
Rosenbaum DM, Rasmussen SGF, Kobilka BK (2009) The structure and function of G-protein-
coupled receptors. Nature 459:356–363
Saengsawang W, Rasenick MM (2015) G protein-coupled receptors. In: Encyclopedia of cell
biology. Academic Press, Cambridge, pp 51–55
Sansom MSP, Adcock C, Smith GR (1998) Modelling and simulation of ion channels: applications
to the nicotinic acetylcholine receptor. J Struct Biol 121:246–262
Schwartz TW, Holst B (2007) Allosteric enhancers, allosteric agonists and ago-allosteric
modulators: where do they bind and how do they act? Trends Pharmacol Sci 28:366–373
Shaik K, Deb PK, Mailavaram RP et al (2019) 7-Amino-2-aryl/hetero-aryl-5-oxo-5,8-dihydro
[1,2,4]triazolo[1,5-a]pyridine-6-carbonitriles: synthesis and adenosine receptor binding studies.
Chem Biol Drug Des 94:1568–1573
Shuker SB, Hajduk PJ, Meadows RP, Fesik SW (1996) Discovering high-affinity ligands for
proteins: SAR by NMR. Science (80-) 274:1531–1534
Stephenson RP (1956) A modification of receptor theory. Br J Pharmacol Chemother 11:379–393
Suvarna BS (2011) Drug - receptor interactions. Kathmandu Univ Med J 9:9–13
Talapatra SK, Tham CL, Guglielmi P et al (2018) Crystal structure of the Eg5 - K858 complex and
implications for structure-based design of thiadiazole-containing inhibitors. Eur J Med Chem
156:641–651
Tallarida RJ (2007) Interactions between drugs and occupied receptors. Pharmacol Ther
113:197–209
Topiol S, Sabio M (2009) X-ray structure breakthroughs in the GPCR transmembrane region.
Biochem Pharmacol 78:11–20
Uings IJ, Farrow SN (2000) Cell receptors and cell signalling. Mol Pathol 53:295–299
Varma AK, Patil R, Das S et al (2010) Optimized hydrophobic interactions and hydrogen bonding
at the target-ligand interface leads the pathways of drug-designing. PLoS One 5:e12029
Wilson DK, Rudolph FB, Quiocho FA (1991) Atomic structure of adenosine deaminase complexed
with a transition-state analog: understanding catalysis and immunodeficiency mutations. Sci-
ence 252:1278–1284
68 B. Chandrasekaran et al.
Wolfenden R, Kati WM (1991) Testing the limits of protein-ligand binding discrimination with
transition-state analogue inhibitors. Acc Chem Res 24:209–215
Zhang T, Wei T, Han Y et al (2016) Protein-ligand interaction detection with a novel method of
transient induced molecular electronic spectroscopy (TIMES): experimental and theoretical
studies. ACS Cent Sci 2:834–842
Pharmacology of Acetylcholine
and Cholinergic Receptors 3
Sarah Falah Kokaz, Pran Kishore Deb, Sara Nidal Abed, Amal Al-
Aboudi, Nirupam Das, Fatimah Amin Younes, Ruba Anwar Salou,
Yazan A. Bataineh, Katharigatta N. Venugopala,
and Raghu Prasad Mailavaram
Abstract
Alzheimer’s disease. Alongside, they are also investigated for various promising
therapeutics. This chapter provides an overview of the cholinergic system phar-
macology, functions in the body, cholinergic and anticholinergic compounds, and
their potential role in the medical field. Further, the chapter highlights the updates
on the cholinergic compounds currently used to treat various conditions as well as
compounds under investigation.
Keywords
Acetylcholine · Cholinergic system · Nicotinic receptors · Muscarinic receptors ·
Acetylcholinesterase inhibitors · Cholinergic ligands
Abbreviation
ACh Acetylcholine
AChE Acetylcholinesterase
AD Alzheimer’s disease
ADHD Attention deficit hyperactivity disorder
ANS Autonomic nervous system
ASDs Autism spectrum disorders
BQCA Benzyl quinolone carboxylic acid+
BTX Botulinum toxin
BuChE Butyrylcholinesterase
ChAT Choline acetyltransferase
ChE Cholinesterase
CNS Central nervous system
CoA Coenzyme A
COPD Chronic obstructive pulmonary disease
DMN Default mode network
DS Down syndrome
ECT Electroconvulsive therapy
EDHF Endothelium-derived hyperpolarizing factor
EDRF Endothelium-derived relaxing factors
EPSP Excitatory postsynaptic potential
FDA Food and Drug Administration
GPCR G-protein-coupled receptor
LAMA Long-acting muscarinic receptor antagonist
mAChR Muscarinic acetylcholine receptor
MCI Mild cognitive impairment
MTL Medial temporal lobe
nAChR Nicotinic acetylcholine receptor
NAL Neutral allosteric ligand
NAM Negative allosteric modulator
NMJ Neuromuscular junction
NO Nitric oxide
3 Pharmacology of Acetylcholine and Cholinergic Receptors 71
3.1 Introduction
disease, and glaucoma (Potter and Kerecsen 2017). Nicotinic receptors stimulation
has shown to improve cognitive functioning after several studies on animal models.
Besides, some preclinical and clinical studies suggested that nicotinic receptors also
play a role in depression, mood, and anxiety (Aboul-Fotouh 2015; Quik et al. 2015).
In this chapter, we present an overview of the cholinergic system pharmacology,
functions in the body, cholinergic and anticholinergic compounds, and their role in
the medical field. Finally, an update on the cholinergic compounds currently used to
treat various conditions as well as compounds under investigation are discussed.
Acetylcholine (ACh)
Acetylcholine (ACh)
Fig. 3.2 Synthesis of acetylcholine by choline acetyltransferase (ChAT) using choline and acetyl
coenzyme A, releasing coenzyme A (HS-CoA). Metabolism of acetylcholine is catalyzed by
acetylcholinesterase (AChE) producing acetic acid and choline
3 Pharmacology of Acetylcholine and Cholinergic Receptors 73
Acetylcholine (ACh) is a neurotransmitter that functions in both the PNS and the
CNS. The ANS (sympathetic and parasympathetic) uses acetylcholine to generate a
nerve impulse. In PNS, ACh mainly acts on the muscular system by activating
muscle contraction after being released in the neuromuscular junction. In the central
nervous system, ACh has various effects on cognitive functions, alertness, learning,
and memory (Haerter and Eikermann 2016; Maurer and Williams 2017; Panus et al.
2009). When ACh gets released from somatic nerve endings into the neuromuscular
junction, it causes the nicotinic ligand-gated ion channels to open, leading to sodium
entrance into muscle cells. The sodium ions produce an excitatory postsynaptic
potential (EPSP) that generates an action potential which eventually stimulates
muscle contraction (Pappano 2018). Also, serum ACh has shown to produce vaso-
dilation by activating vascular endothelial muscarinic receptors, which causes the
release of endothelium-derived relaxing factors (EDRF) including prostanoids,
endothelium-derived hyperpolarizing factor (EDHF), and nitric oxide (Tangsucharit
et al. 2016). The chapter further presents the role of acetylcholine and cholinergic
system together with few specific ligands and their clinical utility.
Cholinergic receptors were named “cholinergic” due to their activation by Ach, and
these receptors are mostly parasympathetic and transduce signal in the autonomic
and somatic nervous system (Wehrwein et al. 2016). Based on the stimulation by
muscarine or nicotine, cholinergic receptors are classified into muscarinic and
nicotinic receptors, respectively. Nicotinic receptors are ionotropic ligand-gated
channels, unlike muscarinic receptors, which are G-protein-coupled receptors
(GPCRs) (Kruse et al. 2014; Papke 2014). Both receptor types are present within
the CNS; however, nicotinic receptors are also found at the neuromuscular junction.
The difference in signal transduction upon activation of the two receptor types
results in distinctive physiological functions.
Muscarinic receptors are mostly present at parasympathetic target organs, but it can
also be found at specific sympathetic target organs such as the eccrine sweat glands
that produce copious secretion for thermoregulation purpose, and in blood vessels of
the skeletal muscles. In the PNS, muscarinic receptors are mainly found on auto-
nomic effector cells, which are innervated by postganglionic parasympathetic nerves
74 S. F. Kokaz et al.
Table 3.2 Muscarinic receptors subtypes localization and role in the brain
Subtype Presence in brain Functional aspect
M1 All major areas of the forebrain Highly responsible for cholinergic
including cerebral cortex, hippocampus, functions. Synaptic plasticity, learning
thalamus, and corpus striatum. Cellular and memory (cognition), neuronal
localization at striatum nigrum neurons differentiation during development, and
and glutamatergic pyramidal neurons neuronal excitability
M2 Throughout the brain especially in Inhibitory effect on dopaminergic action.
hippocampus and neocortex. Abundant Antinociceptive effect reported
in non-cholinergic neurons in these areas
M3 Highly expressed in hypothalamus, Major role in food intake, body growth
lesser expression in hippocampus
M4 Major presence in corpus striatum An important role in psychosis, an
involvement in pathology of Parkinson’s
disease, inhibits D1 receptor of dopamine
signaling
M5 Pars compacta of substantia nigra, Rewarding effect of abusive drugs
ventral tegmental region
(Tiwari et al. 2013). Muscarinic receptors are classified into five subtypes; each
subtype distribution and role are listed in Table 3.1 (Glavind and Chancellor 2011).
These five muscarinic receptor subtypes are expressed throughout the human
brain and are involved in various functional processes, such as learning, memory,
attention, sleep-wake cycles, sensorimotor processing, and arousal (Lebois et al.
2018). Table 3.2 summarizes the details concerning muscarinic receptors distribu-
tion and function in the brain (Verma et al. 2018). Muscarinic receptors are GPCRs
in which M1, M3, and M5 are Gq/11 G-proteins which mediate excitatory
neuromodulatory acetylcholine actions while M2 and M4 receptors are Gi/o
G-proteins that produce inhibitory neuromodulatory acetylcholine actions (Brown
2019; Felder 1995; Lebois et al. 2018).
All five muscarinic receptor subtypes express throughout the mammalian brain.
However, different regions in the brain contain different receptor subtype
concentrations exemplified by M1 and M2 receptors. In the major forebrain areas,
3 Pharmacology of Acetylcholine and Cholinergic Receptors 75
Darifenacin Tolterodine
2017). Such adverse effects of muscarinic receptor antagonists have reduced their
convenience as treatment options; nevertheless they are highly active compounds
(Korczynska et al. 2018) with scope for further structural optimization.
In CNS disorders, the majority of studies are focused on compounds targeting M1
and M4 mAChRs subtypes. On the other hand, M5 mAChRs expresses itself in
highly variable regions of therapeutic interest, and it is the least mAChR expressed
in the brain by 2% of the total mAChRs population. Despite the fact, M5 receptors
are the only subtype with an identifiable mRNA transcript in dopamine-containing
neurons. This factor makes it a viable target for drug addiction treatment by
supporting the hypothesis that mAChR subtype may be responsible for regulating
the transmission of midbrain dopamine and reward mechanisms (Berizzi et al. 2016;
Gunter et al. 2018).
with AD (Congreve et al. 2018; Sivaraman et al. 2019). Figure 3.4 depicts the
chemical structures of some mAChR agonists mentioned in this section.
Bethanechol is a cholinergic agonist that acts on M1 and M2 muscarinic
receptors. It is a methyl analogue of acetylcholine and used clinically for women
with underactive bladder (UAB). Stimulation of mAChRs at the neuromuscular
junction of smooth muscle with bethanechol can induce the contraction of the
detrusor muscle, which eventually improves bladder emptying. However, various
clinical evidence supporting bethanechol efficacy for UAB is limited and poor
(Sivaraman et al. 2019; Gaitonde et al. 2018). Bethanechol has also been
investigated in the treatment of tracheomalacia. However, further trials are required
to ensure the safety and efficacy of bethanechol as a treatment regimen (Bass et al.
2018).
BuTAC ([5R-(exo)]-6-[4-butylthio-1,2,5-thiadiazol-3-yl]-1-azabicyclo-[3.2.1]-
octane) is a muscarinic receptor agonist that produces full agonist activity on M2
receptor and partial agonist activity on both M1 and M4 receptors. Contrarily, it
exhibits full antagonist activity on M3 and M5 receptors. BuTAC has shown to
produce antipsychotic activity on schizophrenic animal models (Andersen et al.
2015; Watt et al. 2013). Another muscarinic agonist is cevimeline, which activates
both M1 and M3 receptors, and it was originally developed for the treatment of
Alzheimer’s-type senile dementia. Currently, this compound is used to treat
xerostomia, which is a condition that causes numerous disorders in oral functions,
including swallowing, taste, speech, and mastication. Other than pilocarpine,
cevimeline is the only agent available for the therapeutic intervention of xerostomia
(Kishimoto et al. 2016; Mitoha et al. 2017). Table 3.5 summarizes various
80 S. F. Kokaz et al.
Table 3.6 Muscarinic receptor antagonists that are currently used and their therapeutic
applications
Agent Use
Tertiary amines
Atropine Treatment of anticholinergic poisoning
Scopolamine Treatment of motion sickness
Homatropine Mydriatic and cycloplegic; for mild uveitis
Dicyclomine Alleviates GI spasms, pylorospasm, and biliary distention
Darifenacin Treatment of urinary incontinence
Fesoterodine Treatment of urinary incontinence
Oxybutynin Treatment of urinary incontinence
Tolterodine Treatment of urinary incontinence
Oxyphencyclimine Antisecretory agent for peptic ulcer
Cyclopentolate Mydriatic and cycloplegic
Tropicamide Mydriatic and cycloplegic
Benztropine Treatment of Parkinson’s and Huntington’s diseases
Trihexyphenidyl Treatment of Parkinson’s and Huntington’s diseases
Pirenzepine Antisecretory agent for peptic ulcer
Quaternary ammonium derivatives
Methylatropine Mydriatic, cycloplegic, and antispasmodic
Methylscopolamine Antisecretory agent for peptic ulcer, antispasmodic
Ipratropium Aerosol for COPD
Glycopyrrolate Antisecretory agent for peptic ulcer, antispasmodic
Tolterodine Treatment of urinary incontinence
Propantheline GI antispasmodic
Tiotropium Aerosol for COPD
a potent and selective M1 PAM, is under preclinical and clinical trials to reduce
cognitive dysfunction in AD patients (Uslaner et al. 2018).
The nAChRs are a part of the “Cys-loop” receptor superfamily, which are multi-
subunit transmembrane receptors. nAChRs mediate excitatory neurotransmission in
autonomic ganglia, specific synapses in both the spinal cord and brain, and at the
vertebrate neuromuscular junction (Albuquerque et al. 2009; Alcaino et al. 2017).
These nAChRs generate complex calcium signals that influence neuronal processes
and signaling molecules. The combination of the nicotinic receptor subtype, Ca2+
signaling pathway, neuronal type, and developmental stage all together translate the
stimulation of the nicotinic receptors into a neuronal response and eventually into a
physiological outcome (Dajas-Bailador and Wonnacott 2004; Kabbani and Nichols
2018). As stated before, nicotinic receptors are ligand-gated ion channels, which are
pentamers assembled from five subunits either as homomeric or heteromeric
pentamers (Fig. 3.5) (Improgo et al. 2010; Millar and Gotti 2009; Wu et al. 2016).
α7 α7 β4 β4
During the inactivated state, the pentamers exist as a funnel shape structural motif
with a central core, and it remains closed. When activated by ACh, the channel
undergoes a conformational change in all subunits, and the core opens to allow Na+
and K+ flow according to electrochemical gradients leading to cellular response
(Svorc 2018). The nAChRs in the CNS are responsible for regulating various
processes including neuronal integration, neurotransmitter release, and cell
excitability and can also affect certain physiological functions including cognition,
pain, arousal, sleep, and mood (Rahman et al. 2015).
Nicotine, a prototypic tobacco alkaloid, had received interest as a potential
therapeutic compound 30 years ago, which was the time when tobacco smoking
was discovered to affect the performance of a particular cognitive task positively.
Further investigations substantiated that nicotine administration indeed affects
pro-cognitive performances (Terry and Callahan 2018; Heishman et al. 1994;
Sherwood 1993). Nowadays, growing pieces of evidence corroborate nicotine’s
effect in enhancing cognitive function and information processing in both experi-
mental animals and human nonsmokers (Levin et al. 2013). Additionally, other
effects of nicotine have been reported from in vivo and in vitro studies such as
sustained attention, working memory (Newhouse 2019) and recognition memory in
rats (Nikiforuk et al. 2015), attention, processing of visual information, and short
term memory in humans. In vivo and in vitro studies have also shown that nicotine
possesses neuroprotective activity in specific disease models which proposes that
nicotine can be used not only for symptomatic control but also to produce effects that
enhance cognition in neurodegenerative diseases such as AD and PD (Newhouse
2019). However, nicotine has limited therapeutic potentials for psychiatric and
neurologic conditions due to several factors including cardiovascular side effects,
abuse potentials, and its relatively short half-life. Nicotine transdermal patches used
for smoking cessation have shown no cardiovascular side effects while exhibiting
improved cognition in patients with mild cognitive impairment (MCI) (Newhouse
et al. 2012). Various types of nicotine formulations have been developed for
smoking cessation such as tablets, patches, nasal sprays, gums, inhalers, and
lozenges to reduce cravings and withdrawal symptoms (Shahab et al. 2013). Nico-
tinic acetylcholine receptor ligands other than nicotine have various therapeutic
potentials which are listed in Fig. 3.6 (Terry and Callahan 2018).
Nicotinic cholinergic receptors (nAChRs) are pentameric proteins that are
activated by acetylcholine at central and peripheral synapses. Neuronal nicotinic
receptors in the brain, autonomic ganglia, adrenal gland, and immune cells are
composed of either α subunits or both α and β subunits. One of the primary
phenomena that nicotinic receptors exhibit upon continued nicotinic agonist expo-
sure is desensitization, which occurs in both muscular and neuronal nicotinic
receptors.
Nicotine
Dependence
Therapeutic
Parkinson’s Potential of
Depression
Disease nAChR
Ligands
Attention-
Deficit/Hype Alzheimer’s
ractivity Disease
Disorder
Schizophrenia
Fig. 3.6 Potential therapeutic applications of nicotinic acetylcholine receptors (nAChRs) ligands
Lobeline Nicotine
Cytisine Varenicline
agonists started in the early 1990s. ABT-418 was one of the first nicotinic agonists
developed by Abbott Labs. Further investigations of ABT-418 have shown
cognitive-enhancing effects that are three to ten times more potent than nicotine.
Clinical trials on ADHD patients with ABT-418 have shown promising results in
3 Pharmacology of Acetylcholine and Cholinergic Receptors 87
There are two types of cholinesterases in the body, acetylcholinesterase (AChE) and
butyrylcholinesterase (BuChE), which differ in substrate specificity and their distri-
bution within the body. While AChE terminates the action of ACh at synapses in the
nervous system, BuChE concentrates in non-neuronal sites such as liver and plasma.
BuChE is also responsible for metabolizing certain drugs (e.g., ester-type local
anesthetics, succinylcholine) (Colovic et al. 2013; Kishore et al. 2012; Mehrpouya
et al. 2017).
Most compounds used clinically to inhibit cholinesterase work on both the
enzymes without discrimination. Cholinesterase inhibitors are classified into two
main types based on the nature of the compound-enzyme binding, viz. reversible and
irreversible. Reversible compounds can either be covalent or non-covalent
inhibitors. Edrophonium is a non-covalent inhibitor that binds reversibly to the
anionic part of the acetylcholinesterase enzyme. Drug-enzyme bond characteristics
mainly determine the duration of these compounds. For instance, edrophonium binds
weakly to the enzyme and is rapidly cleared by the kidney, which results in a short
duration of action that is approximately 10 min long (Romano et al. 2017; dos
Coelho et al. 2018). Other examples of non-covalent ChE inhibitors are donepezil
and tacrine used for the treatment of AD. These two compounds possess a longer
duration of action due to their partition into lipids and their higher affinities (Birks
and Harvey 2018; Sameem et al. 2017). Carbamic acid ester derivatives such as
neostigmine and physostigmine are covalent inhibitors, sometimes termed as carba-
mate inhibitors. After their binding to the AChE enzyme, they get hydrolyzed, which
results in carbamylation of the serine residue in the active site of the enzyme (Arens
and Kearney 2019; Lauretti 2015).
On the other hand, irreversible ChE inhibitors act on the serine group in the active
site of the enzyme and phosphorylate it. Nerve gases such as tabun, sarin, and soman
are examples of irreversible ChE inhibitors. Other examples include insecticides
(malathion, parathion) and therapeutic agents such as isofluorophate and
echothiophate referred to as organophosphate or organophosphorus ChE inhibitors.
The reason why these compounds are considered irreversible is that the resultant
phosphorylated enzyme is exceptionally stable, which means that the dephosphory-
lation process can take hours to occur. The possibility of dephosphorylation of
phosphorylated enzyme by secondary and tertiary alkyl-substituted phosphates
(soman and isofluorophate) cannot be achieved to restore the enzyme activity until
further biosynthesis of new enzyme molecules (Sánchez-Santed et al. 2016; Thapa
et al. 2017).
Inhibition of ChE enzyme by anticholinesterase results in augmentation of
acetylcholine and the effects get relayed throughout the whole nervous system
leading to both therapeutic actions and associated adverse effects. Hence those
compounds lead to a nonselective indirect activation of both muscarinic and nico-
tinic receptors. Therefore, the receptors in the peripheral nervous system and the
brain get activated, including peripheral tissues innervated by parasympathetic
nerves, sympathetic and parasympathetic ganglia, and at the neuromuscular junction
90 S. F. Kokaz et al.
(Ehlert 2019b). Many acetylcholinesterase inhibitors are available on the market for
the treatment of various conditions (Table 3.7) (Potter and Kerecsen 2017).
Release inhibitors’ primary mechanism of action is preventing ACh release from the
presynaptic end of the neuron. Botulinum toxin (BTX) is produced by Clostridium
botulinum bacteria that target cholinergic receptors at neuromuscular junctions in
skeletal muscles, inhibiting ACh release leading to neuromuscular blockage and
paralysis. The inhibition of ACh occurs after the toxin enters the nerve and reaches
the cytoplasm, then cleaves the (SNARE) proteins that are responsible for vesicle
fusion mediation. Prevention of vesicle fusion means that the ACh vesicles can no
longer bind into the intracellular cell membrane and release ACh into the synaptic
cleft (Dressler and Saberi 2005; Zhao et al. 2016). Some snake venoms produce an
irreversible blockade of neuromuscular transmission such as Crotoxin (CTX). CTX
is a heterodimeric phospholipase A2 (PLA2) neurotoxin produced by a Brazilian
rattlesnake (Crotalus durissus terrificus). This neurotoxin causes prevention of
acetylcholine release from presynaptic ends, which depends on intrinsic PLA2
activity (Cavalcante et al. 2017). Blockage of ACh release leads to interruption of
neuromuscular transmission and eventual muscle paralysis. Crotoxin exerts postsyn-
aptic level effects too by stabilizing desensitized acetylcholine receptors (Faure et al.
2017).
Table 3.7 Currently used acetylcholinesterase inhibitors (AChEIs) for various therapeutic applications
CNS
Drug Brand name Class/type of binding Duration effect Uses
Physostigmine Eserine Carbamate, reversible 0.5–2 h Yes Treatment of atropine poisoning, glaucoma
Neostigmine Prostigmin Carbamate, reversible 0.5–4 h No Reversal of NMJ blockade, myasthenia gravis
Pyridostigmine Mestinon, Carbamate, reversible 4–6 h No Reversal of NMJ blockade, myasthenia gravis, prevention of
Regonol organophosphate poisoning
Ambenonium Mytelase Carbamate, reversible 3–8 h No Reversal of NMJ blockade, myasthenia gravis
Tacrine Cognex Pyridine, reversible 4–6 h Yes Alzheimer’s disease (discontinued)
Donepezil Aricept Piperidine, non-covalent, >6 h Yes Yes Alzheimer’s disease
reversible
Galantamine Reminyl, Tertiary alkaloid, >6 h Yes Yes Alzheimer’s disease
Razadyne reversible
Rivastigmine Exelon Carbamate, pseudo- 10–12 h Yes Alzheimer’s disease
Pharmacology of Acetylcholine and Cholinergic Receptors
irreversible
Edrophonium Tensilon Electrostatic, rapidly 5–15 min No Diagnosis of myasthenia gravis, reversal of NMJ blockade
reversible
Echothiophate Phospholine Organophosphate, >24 h N/A Glaucoma
irreversible (topical)
91
92 S. F. Kokaz et al.
Nicotinic receptor
stimulation
Efficiency in
DMN activity in
attention and
frontal, cingulate
executive function
& parietal regions
networks
Attention
Distraction
Encoding of relevant info
Processing speed
Enhanced memory,
Planning and functioning
Fig. 3.8 Simplified model of nicotinic receptor stimulation effect in the enhancement of cognitive
functioning. Abbreviations: MTL: medial temporal lobe; DMN: default mode network
altogether may enhance processing speed, attention, and response inhibition, even-
tually leading to improved memory and attention to external stimuli and decreased
internally focused processing (Hahn 2019; Newhouse 2019).
Nicotine and nAChRs ligands have shown a different level of enhancement in
cognitive functions after many studies carried out on animal models, including
attention, memory, and learning. Further clinical trials have shown that these
cognitive effects are also present when varenicline has improved cognitive
impairment in schizophrenic patients and increased lapses in attention in smokers.
Recent clinical trials have shown that activating α7 nAChRs has procognitive effects
on patients with AD and schizophrenia. Likewise, a randomized placebo-controlled
double-blinded study with TC-1734, a β2-selective nicotinic agonist, had shown an
improved cognition when given to patients with age-associated memory impairment
(Hoskin et al. 2019; Quik et al. 2015). Some preclinical and clinical studies
supported that nicotinic receptors have a role in anxiety, mood, and depression. A
study revealed that the treatment of Tourette’s disorder with mecamylamine, a
nAChR antagonist, showed considerable improvement in depression symptoms
(Aboul-Fotouh 2015; Silver et al. 2001).
Nicotinic receptors are the principal receptors affecting the regulatory pathways
of nicotinic and cholinergic signaling, as suggested by recent studies. These signals
3 Pharmacology of Acetylcholine and Cholinergic Receptors 93
Alzheimer's
disease therapy
AChE NMDA
inhibitors antagonist
Tacrine
(withdrawn)
Galantamine Rivastigmine Donepezil Memantine
dysfunction in those patients increases the risk of falls and hospitalizations. 80% of
PD patients develop dementia after 20 years, which makes dementia the leading risk
factor for falls. Treatments that interfere with both dopaminergic and cholinergic
imbalance can improve long-term outcomes of PD patients (Hiller et al. 2015;
Pagano et al. 2015).
Organophosphorus compounds (OPCs) are primarily a group of pesticides that
inhibit acetylcholinesterase, and they are considered a severe health hazard. Current
therapy for OPCs exposure is oxime-type enzyme reactivators such as obidoxime
and pralidoxime, while atropine is administered for symptomatic treatment some-
times with unsatisfactory therapeutic outcomes. Better therapeutic results are
observed when AChE inhibitors (reversible) are administered before the exposure
to organophosphorus compounds. Recent studies showed that bispyridinium oximes
K027 and K203, AChE inhibitors under development, are highly effective (K027
more than K203) in protecting against OPCs when administered pre-exposure. This
strategy makes it a promising prophylactic agent and preventing possible occupa-
tional hazard (Antonijevic et al. 2016; Lorke and Petroianu 2018).
3.5 Conclusion
Cholinergic ligands possess great importance for various medical conditions. Over
the years, studies and research have provided substantial evidence of positive
outcomes from cholinergic ligands, such as nicotinic stimulators and muscarinic
agonist and their ability to improve cognitive performance. Currently, there are
potential cholinergic ligands approved for medical applications such as acetylcho-
linesterase (AChE) inhibitors including galantamine, donepezil, and rivastigmine for
the treatment of AD. Furthermore, several AChE inhibitors displayed promising and
appreciable therapeutic benefit for glaucoma, atropine poisoning, myasthenia gravis,
and reversal of NMJ (neuromuscular junction) blockade. Another example is mus-
carinic receptor antagonists (tertiary amines, quaternary ammonium derivatives)
used for conditions such as urinary incontinence, motion sickness, GI antispas-
modic, COPD, Parkinson’s disease, Huntington’s disease, and as an antisecretory
agent for peptic ulcer. The involvement and pathophysiology of cholinergic
receptors in certain medical conditions are subsequently investigated to provide
more potential therapeutic targets in the future. For example, α9-nAChR
upregulation in estrogen-positive breast cancer suggests that nicotinic receptors
can mediate oncogenic signaling, making them a potential new target in treating
and preventing certain cancers. Gene knockout technology is currently being used to
provide a further understanding of the cholinergic receptors, especially in the brain
to present new potential therapeutic agents for various conditions.
96 S. F. Kokaz et al.
References
Aarsland D, Creese B, Politis M, Chaudhuri KR, Ffytche DH, Weintraub D, Ballard C (2017)
Cognitive decline in Parkinson disease. Nat Rev Neurol 13:217–231
Abdel-Magid AF (2015) Allosteric modulators: an emerging concept in drug discovery. ACS Med
Chem Lett 6:104–107
Aboul-Fotouh S (2015) Behavioral effects of nicotinic antagonist mecamylamine in a rat model of
depression: prefrontal cortex level of BDNF protein and monoaminergic neurotransmitters.
Psychopharmacology 232:1095–1105
Akaike A, Izumi Y (2018) Overview. In: Akaike A, Shimohama S, Misu Y (eds) Nicotinic
acetylcholine receptor signaling in Neuroprotection. Springer, New York, NY, pp 1–16
Albuquerque EX, Pereira EFR, Manickavasagom Alkondon SWR (2009) Mammalian nicotinic
acetylcholine receptors: from structure to function. Physiol Rev 89:73–120
Alcaino C, Musgaard M, Minguez T, Mazzaferro S, Faundez M, Iturriaga-Vasquez P, Biggin PC,
Bermudez I (2017) Role of the Cys loop and transmembrane domain in the allosteric modulation
of α4β2 nicotinic acetylcholine receptors. J Biol Chem 292:551–562
Aleem A, Janbaz KH (2018) Involvement of dual mechanisms of anti-muscarinic and Ca++
antagonistic activities to validate the folkloric uses of Cyperus niveus Retz. in multiple gastro-
intestinal ailments. J Ethnopharmacol 213:138–148
Alvarado-Gonzalez A, Arce I (2015) Tiotropium bromide in chronic obstructive pulmonary disease
and bronchial asthma. J Clin Med Res 7:831–839
Andersen MB, Croy CH, Dencker D, Werge T, Bymaster FP, Felder CC, Fink-Jensen A (2015)
Antipsychotic-like effect of the muscarinic acetylcholine receptor agonist BuTAC in
non-human Primates. PLoS One 10:1–13
Andersson K-E (2019) Future considerations in overactive bladder pharmacotherapy. In: Cox L,
Rovner ES (eds) Contemporary pharmacotherapy of overactive bladder. Springer, New York,
NY, pp 219–229
Antonijevic E, Musilek K, Kuca K, Djukic-Cosic D, Vucinic S, Antonijevic B (2016) Therapeutic
and reactivating efficacy of oximes K027 and K203 against a direct acetylcholinesterase
inhibitor. Neurotoxicology 55:33–39
Arens AM, Kearney T (2019) Adverse effects of physostigmine. J Med Toxicol 15(3):184–191
Bass R, Santiago M, Smith L, Quinlan C, Panitch H, Giordano T, Piccione J (2018) Bethanechol in
Tracheomalacia: two case series and a review of the literature. Pediatr Allergy Immunol
Pulmonol 31:180–183
Bender AM, Jones CK, Lindsley CW (2017) Classics in chemical neuroscience : Xanomeline. ACS
Chem Neurosci 8:435–443
Bender AM, Garrison AT, Lindsley CW (2019) The muscarinic acetylcholine receptor M5:
therapeutic implications and allosteric modulation. ACS Chem Neurosci 10:1025–1034
Berizzi AE, Gentry PR, Rueda P, Den Hoedt S, Sexton PM, Langmead CJ, Christopoulos A (2016)
Molecular mechanisms of action of M5 muscarinic acetylcholine receptor allosteric modulators.
Mol Pharmacol 90:427–436
Berizzi AE, Perry CJ, Shackleford DM, Lindsley CW, Jones CK, Chen NA, Sexton PM,
Christopoulos A, Lawrence CJLAJ (2018) Muscarinic M5 receptors modulate ethanol seeking
in rats. Neuropsychopharmacology 43:1510–1517
Bertrand D Jr, Terry AV (2018) The wonderland of neuronal nicotinic acetylcholine receptors.
Biochem Pharmacol 151:214–225
Bhat SK, Ashwin D, Mythri S, Bhat S (2017) Arecanut (Areca catechu L) decreases Alzheimer’s
disease symptoms: compilation of research works. J Med Plants Stud 5:4–9
Birks JS, Harvey RJ (2018) Donepezil for dementia due to Alzheimer’s disease. Cochrane Database
Syst Rev 6:1465–1858
Blessing B, Gibbins I (2016) Autonomic nervous system. In: Prescott TJ, Ahissar E, Izhikevich E
(eds) Scholarpedia of touch. Springer, New York, NY, pp 467–477
3 Pharmacology of Acetylcholine and Cholinergic Receptors 97
D’Souza RS, Johnson RL (2019) Nondepolarizing paralytics [WWW document]. StatPearls Publ,
Treasure Island, FL
Dajas-Bailador F, Wonnacott S (2004) Nicotinic acetylcholine receptors and the regulation of
neuronal signalling. Trends Pharmacol Sci 25:317–324
Dang N, Meng X, Song H (2016) Nicotinic acetylcholine receptors and cancer (review). Biomed
Rep 4:515–518
Dani JA (2015) Neuronal nicotinic acetylcholine receptor structure and function and response to
nicotine. Int Rev Neurobiol 124:3–19
de Linder Henriksen M, Lim C, Sharp JL (2019) Treatment of canine postoperative ocular
hypertension with combined latanoprost 0.005% and atropine 1% ophthalmic solutions. Vet
Ophthalmol 22(4):448–461
de Souza LG, Rennó MN, Figueroa-Villar JD (2016) Coumarins as cholinesterase inhibitors: a
review. Chem Biol Interact 254:11–23
Deutsch SI, Burket JA, Benson AD (2014) Targeting the α7 nicotinic acetylcholine receptor to
prevent progressive dementia and improve cognition in adults with Down’s syndrome. Prog
Neuro-Psychopharmacol Biol Psychiatry 54:131–139
Deutsch SI, Burket JA, Urbano MR, Benson AD (2015) The α7 nicotinic acetylcholine receptor: a
mediator of pathogenesis and therapeutic target in autism Spectrum disorders and down
syndrome. Biochem Pharmacol 97:363–377
Digby GJ, Shirey K, Conn PJ (2010) Allosteric activators of muscarinic receptors as novel
approaches for treatment of CNS disorders. Mol BioSyst 6:1345–1354
Digby GJ, Noetzel MJ, Bubser M, Utley TJ, Walker AG, Byun NE, Lebois EP, Xiang Z, Sheffler
DJ, Cho HP, Davis AA, Nemirovsky NE, Mennenga SE, Camp BW, Bimonte-Nelson HA,
Bode J, Italiano K, Morrison R, Daniels JS, Niswender CM, Olive MF, Lindsley CW, Jones CK,
Conn PJ (2012) Novel allosteric agonists of M1 muscarinic acetylcholine receptors induce brain
region-specific responses that correspond with behavioral effects in animal models. J Neurosci
32:8532–8544
Dineley KT, Pandya AA, Yakel JL (2015) Nicotinic ACh receptors as therapeutic targets in CNS
disorders. Trends Pharmacol Sci 36:96–108
Ding X-Q (2017) Drugs affecting nicotinic receptors. In: Dowd FJ, Johnson BS, Mariotti AJ (eds)
Pharmacology and therapeutics for dentistry. Elsevier, Amsterdam, pp 98–109
dos Coelho ST, Gomes TM, Serra PBA, Camara AL, Paes AMA (2018) Naturally occurring
Acetylcholinesterase inhibitors and their potential use for Alzheimer’s disease therapy. Front
Pharmacol 9:1–14
Dressler D, Saberi FA (2005) Botulinum toxin: mechanisms of action. Eur Neurol 53:3–9
Drudi FM, Lima C, Freitas L, Yogi M, Nascimento H, Junior RB (2017) Acetylcholine chloride 1%
usage for intraoperative cataract surgery miosis. Rev Bras Oftalmol 76:247–249
Egea J, Buendia I, Parada E, Navarro E, León R, Lopez MG (2015) Anti-inflammatory role of
microglial alpha7 nAChRs and its role in neuroprotection. Biochem Pharmacol 97:463–472
Ehlert F (2019a) Muscarinic antagonists and their clinical uses. In: Wecker L (ed) Brody’s human
pharmacology mechanism-based therapeutics. Elsevier, Amsterdam, pp 268–282
Ehlert F (2019b) Muscarinic agonists, Cholinesterase inhibitors, and their clinical uses. In: Wecker
L (ed) Brody’s human pharmacology mechanism-based therapeutics. Elsevier, Amsterdam, pp
241–267
Engers DW, Melancon BJ, Gregro AR, Bertron JL, Bollinger SR, Felts AS, Konkol LC, Wood MR,
Bollinger KA, Luscombe VB, Rodriguez AL, Jones CK, Bubser M, Yohn SE, Wood MW,
Brandon NJ, Dugan ME, Niswender CM, Conn PJ, Bridges TM, Lindsley CW (2019)
VU6005806/AZN-00016130, an advanced M4 positive allosteric modulator (PAM) profiled
as a potential preclinical development candidate. Bioorg Med Chem Lett 29:1714–1718
Fasoli F, Gotti C (2015) Structure of neuronal nicotinic receptors. In: Balfour DJK, Munafò MR
(eds) The neurobiology and genetics of nicotine and tobacco, Current topics in behavioral
neurosciences, vol 23. Springer, New York, NY, pp 1–17
3 Pharmacology of Acetylcholine and Cholinergic Receptors 99
Faure G, Porowinska D, Saul F (2017) Crotoxin from Crotalus durissus terrificus and crotoxin-
related proteins: structure and function relationship. In: Gopalakrishnakone P, Cruz LJ, Luo S
(eds) Toxins and drug discovery. Springer, New York, NY, pp 1–18
Felder C (1995) Muscarinic acetylcholine receptors: signal transduction through multiple effectors.
FASEB J 9:619–625
Ferreira-vieira TH, Guimaraes IM, Silva FR, Ribeiro FM (2016) Alzheimer’s disease: targeting the
cholinergic system. Curr Neuropharmacol 14:101–115
Fisher A (2012) Cholinergic modulation of amyloid precursor protein processing with emphasis on
M1 muscarinic receptor: perspectives and challenges in treatment of Alzheimer’s disease. J
Neurochem 120:22–33
Foster DJ, Conn PJ (2017) Allosteric modulation of GPCRs: new insights and potential utility for
treatment of schizophrenia and other CNS disorders. Neuron 94:431–446
Foster DJ, Choi DL, Conn PJ, Rook JM (2014) Activation of M1 and M4 muscarinic receptors as
potential treatments for Alzheimer’s disease and schizophrenia. Neuropsychiatr Dis Treat
10:183–191
Gaitonde S, Malik RD, Christie AL, Zimmern PE (2018) Bethanechol: is it still being prescribed for
bladder dysfunction in women? Int J Clin Pract 73:e13248. 1–6
Gandelman JA, Kang H, Antal A, Albert K, Boyd BD, Conley AC, Newhouse P, Taylor WD
(2018) Transdermal nicotine for the treatment of mood and cognitive symptoms in non-smokers
with late-life depression. J Clin Psychiatry 79:1–17
Garlapati K, Kammari A, Badam RK, Surekha BE, Boringi M, Soni P (2019) Meta-analysis on
pharmacological therapies in the management of xerostomia in patients with Sjogren’s syn-
drome. Immunopharmacol Immunotoxicol 41:1–7. https://doi.org/10.1080/08923973.2019.
1593448
Gault LM, Lenz RA, Ritchie CW, Meier A, Othman AA, Tang Q, Berry S, Pritchett Y, Robieson
WZ (2016) ABT-126 monotherapy in mild-to-moderate Alzheimer’s dementia: randomized
double- blind, placebo and active controlled adaptive trial and open-label extension. Alzheimers
Res Ther 8:1–13
Gentry PR, Kokubo M, Bridges TM, Kett NR, Harp JM, Cho HP, Smith E, Chase P, Hodder PS,
Niswender CM, Daniels JS, Conn PJ, Wood MR, Lindsley CW (2013) Discovery of the first
M5-selective and CNS penetrant negative allosteric modulator (NAM) of a muscarinic acetyl-
choline receptor: (S)-9b-(4-Chlorophenyl)-1-(3,4-difluorobenzoyl)-2,3-dihydro-1H-imidazo
[2,1-a]isoindol-5(9bH)-one (ML375). J Med Chem 56:9351–9355
Gentry PR, Sexton PM, Christopoulos A (2015) Novel allosteric modulators of G protein-coupled
receptors. J Biol Chem 290:19478–19488
Glavind K, Chancellor M (2011) Antimuscarinics for the treatment of overactive bladder: under-
standing the role of muscarinic subtype selectivity. Int Urogynecol J 22:907–917
Gould RW, Dencker D, Grannan M, Bubser M, Zhan X, Wess J, Xiang Z, Locuson C, Lindsley
CW, Conn PJ, Jones CK (2015) Role for the M1 muscarinic acetylcholine receptor in top-down
cognitive processing using a touchscreen visual discrimination task in mice. ACS Chem
Neurosci 6:1683–1695
Greig NH, Reale M, Tata AM (2013) New advances in pharmacological approaches to the
cholinergic system: an overview on muscarinic receptor ligands and cholinesterase inhibitors.
Recent Pat CNS Drug Discov 8:123–141
Grundey J, Amu R, Ambrus GG, Batsikadze G, Paulus W, Nitsche MA (2015) Double dissociation
of working memory and attentional processes in smokers and non-smokers with and without
nicotine. Psychopharmacology 232:2491–2501
Gunter BW, Gould RW, Bubser M, McGowan KM, Lindsley CW, Jones CK (2018) Selective
inhibition of M5 muscarinic acetylcholine receptors attenuates cocaine self-administration in
rats. Addict Biol 23:1106–1116
Haerter F, Eikermann M (2016) Reversing neuromuscular blockade: inhibitors of the acetylcholin-
esterase versus the encapsulating agents sugammadex and calabadion. Expert Opin
Pharmacother 17:819–833
100 S. F. Kokaz et al.
Hager HH, Burns B (2018) Depolarizing muscle relaxants [WWW document]. StatPearls Publ,
Treasure Island, FL
Hahn B (2019) The therapeutic potential of the cognitive-enhancing effects of nicotine and other
nicotinic acetylcholine receptor agonists. In: Preedy VR (ed) Neuroscience of nicotine. Aca-
demic Press, Cambridge, pp 305–311
Han MK, Lazarus SC (2016) COPD: clinical diagnosis and management. In: Broaddus VC, Mason
RJ, Ernst JD, King TE, Lazarus SC, Murray JF, Nadel JA, Slutsky AS, Gotway MB (eds)
Murray and Nadel’s textbook of respiratory medicine. Elsevier, Amsterdam, pp 767–785
Heishman SJ, Taylor RC, Henningfield JE (1994) Nicotine and smoking: a review of effects on
human performance. Exp Clin Psychopharmacol 2:345–395
Hepnarova V, Korabecny J, Matouskova L, Jost P, Muckova L, Hrabinova M, Vykoukalova N,
Kerhartova M, Kucera T, Dolezal R, Nepovimova E, Spilovska K, Mezeiova E, Pham NL,
Jun D, Staud F, Kaping D, Kuca K, Soukup O (2018) The concept of hybrid molecules of tacrine
and benzyl quinolone carboxylic acid (BQCA) as multifunctional agents for Alzheimer’s
disease. Eur J Med Chem 150:292–306
Hiller A, Nutt J, Mancini M, Horak F, Kareus S, Schoneburg BM, Chung KA (2015) Are
cholinesterase inhibitors effective in improving balance in Parkinson’s disease? J Neurol Disord
2:1–4
Hoskin JL, Al-Hasan Y, Sabbagh MN (2019) Nicotinic acetylcholine receptor agonists for the
treatment of Alzheimer’s dementia: an update. Nicotine Tob Res 21:370–376
Huang L, Miao H, Sun Y, Meng F, Li X (2014) Discovery of indanone derivatives as multi-target-
directed ligands against Alzheimer’s disease. Eur J Med Chem 87:429–439
Improgo MRD, Scofield MD, Tapper AR, Gardner PD (2010) The nicotinic acetylcholine receptor
CHRNA5/A3/B4 gene cluster: dual role in nicotine addiction and lung cancer. Prog Neurobiol
92:212–226
Jakubik J, El-Fakahany EE (2016) Allosteric modulation of muscarinic receptors. In: Myslivecek J,
Jakubik J (eds) Muscarinic receptor: from structure to animal models. Springer, New York, NY,
pp 95–130
Jiang N, Li S-Y, Xie S-S, Li Z-R, Wang KDG, Wang X-B, Kong L-Y (2014) Design, synthesis and
evaluation of multifunctional salphen derivatives for the treatment of Alzheimer’s disease. Eur J
Med Chem 87:540–551
Jones CK, Byun N, Bubser M (2012) Muscarinic and nicotinic acetylcholine receptor agonists and
allosteric modulators for the treatment of schizophrenia. Neuropsychopharmacol Rev 37:16–42
Kabbani N, Nichols RA (2018) Beyond the channel: metabotropic signaling by nicotinic receptors.
Trends Pharmacol Sci 39:354–366
Keating GM (2016) Sugammadex: a review of neuromuscular blockade reversal. Drugs
76:1041–1052
Khatwal RB, Dubala A, Kosaraju J, Chinni S (2014) Pharmacokinetics and tissue distribution of a
M1 muscarinic acetylcholine receptor positive allosteric potentiator , benzyl quinolone carbox-
ylic acid. Anal Methods 6:2672–2678
Kishimoto N, Stegaroiu R, Shibata S, Ito K, Inoue M, Ohuchi A (2016) Changes in the Oral
moisture and the amount of microorganisms in saliva and tongue coating after Oral ingestion
resumption: a pilot study. Open Dent J 10:79–88
Kishore P, Anuradha D, Piplani P, Rao R (2012) Molecular docking and receptor-specific
3D-QSAR studies of acetylcholinesterase inhibitors. Mol Divers 16:803–823
Korczynska M, Clark MJ, Valant C, Xu J, Von Moo E, Albold S, Weissa DR, Torosyan H,
Huang W, Kruse AC, Lyda BR, May LT, Baltos J-A, Sexton PM, Kobilka BK,
Christopoulos A, Shoichet BK, Sunahara RK (2018) Structure-based discovery of selective
positive allosteric modulators of antagonists for the M2 muscarinic acetylcholine receptor.
PNAS 115:2419–2428
Kruse AC, Kobilka BK, Gautam D, Sexton PM, Christopoulos A, Wess J (2014) Muscarinic
acetylcholine receptors: novel opportunities for drug development. Nat Rev Drug Discov
13:549–560
3 Pharmacology of Acetylcholine and Cholinergic Receptors 101
Maurer SV, Williams CL (2017) The cholinergic system modulates memory and hippocampal
plasticity via its interactions with non-neuronal cells. Front Immunol 8:1–14
McGowan KM, Nance KD, Cho HP, Bridges TM, Jones CK, Conn PJ, Jones CK, Lindsley CW
(2017) Continued optimization of the M5 NAM ML375: discovery of VU6008667, an M5
NAM with high CNS penetration and a desired short half-life in rat for addiction studies. Bioorg
Med Chem Lett 27:1356–1359
Mehrpouya M, Ataei S, Nili-Ahmadabadi A (2017) Potential drug interactions with cholinesterase
inhibitors in Alzheimer patients: a guideline for neurologists. J Appl Pharm Sci 7:223–226
Melroy-Greif WE, Stitzel JA, Ehringer MA (2016) Nicotinic acetylcholine receptors: upregulation,
age-related effects and associations with drug use. Genes Brain Behav 15:89–107
Millar NS, Gotti C (2009) Diversity of vertebrate nicotinic acetylcholine receptors. Neuropharma-
cology 56:237–246
Mitoha Y, Ueda H, Ichikawa H, Fujita M, Kobashi M, Matsuo R (2017) Effects of cevimeline on
excitability of parasympathetic preganglionic neurons in the superior salivatory nucleus of rats.
Auton Neurosci 206:1–7
Naicker P, Anoopkumar-Dukie S, Grant GD, Kavanagh JJ (2017) Anticholinergic activity in the
nervous system: consequences for visuomotor function. Physiol Behav 170:6–11
Naser PV, Kuner R (2018) Molecular, cellular and circuit basis of cholinergic modulation of pain.
Neuroscience 387:135–148
Newhouse PA (2019) Therapeutic applications of nicotinic stimulation: successes, failures, and
future prospects. Nicotine Tob Res 21:345–348
Newhouse P, Kellar K, Aisen P, White H, Wesnes K, Coderre E, Pfaff A, Wilkins H, Howard D,
Levin ED (2012) Nicotine treatment of mild cognitive impairment: a 6-month double-blind pilot
clinical trial. Neurology 78:91–101
Nikiforuk A, Kos T, Potasiewicz A, Popik P (2015) Positive allosteric modulation of alpha
7 nicotinic acetylcholine receptors enhances recognition memory and cognitive flexibility in
rats. Eur Neuropsychopharmacol 25:1300–1313
Oba Y, Sarva ST, Dias S (2016) Efficacy and safety of long-acting β-agonist/ long-acting musca-
rinic antagonist combinations in COPD: a network meta-analysis. Thorax 71:15–25
Pagano G, Rengo G, Pasqualetti G, Femminella GD, Monzani F, Ferrara N, Tagliati M (2015)
Cholinesterase inhibitors for Parkinson’s disease: a systematic review and meta-analysis. J
Neurol Neurosurg Psychiatry 86:767–773
Panus PC, Katzung B, Jobst EE, Tinsley SL, Masters SB, Trevor AJ (2009) Drugs affecting the
cholinergic system. In: Pharmacology for the physical therapist. McGraw Hill Professional,
New York, NY, pp 54–70
Papke RL (2014) Merging old and new perspectives on nicotinic acetylcholine receptors. Biochem
Pharmacol 89:1–11
Pappano AJ (2018) Cholinoceptor-activating & cholinesterase-inhibiting drugs. In: Katzung BG
(ed) Basic & clinical pharmacology. McGraw Hill Professional, New York, NY, pp 107–123
Petrakis IL, Ralevski E, Gueorguieva R, O’Malley SS, Arias A, Sevarino KA, Jane JS, O’Brien E,
Krystal JH (2018) Mecamylamine treatment for alcohol dependence: a randomized controlled
trial. Addiction 113:6–14
Picciotto MR, Higley MJ, Mineur YS (2012) Acetylcholine as a neuromodulator: cholinergic
signaling shapes nervous system function and behavior. Neuron 76:116–129
Potter PE, Kerecsen L (2017) Cholinesterase inhibitors. In: Rahman A, Choudhary MI (eds)
Frontiers in CNS drug discovery. Bentham Science, Sharjah, pp 201–239
Quik M, Bordia T, Zhang D, Perez XA (2015) Nicotine and nicotinic receptor drugs: potential for
Parkinson’s disease and drug-induced movement disorders. In: International review of neurobi-
ology. Elsevier, Amsterdam, pp 247–271
Rahman S, Engleman EA, Bell RL (2015) Nicotinic receptor modulation to treat alcohol and drug
dependence. Front Neurosci 8:426
3 Pharmacology of Acetylcholine and Cholinergic Receptors 103
Ramaswamy S, Chhabra SK, Gupta M, Dash DJ, Bansal V (2018) Salbutamol but not ipratropium
shifts autonomic balance towards sympathetic in chronic obstructive pulmonary disease. Curr
Respir Med Rev 14:166–171
Rogers BN, Gray DL (2012) Treating the cognitive deficits of schizophrenia. In: Rankovic Z,
Bingham M, Nestler EJ, Hargreaves R (eds) Drug discovery for psychiatric disorders. RSC
Publishing, Cambridge, pp 123–158
Romano M, Martin-Flores M, Sakai DM, Tseng CT, Campoy L, Gleed RD (2017) Effects of
neostigmine or edrophonium on force of contraction when administered at a train-of-four ratio
of 0.9 in anesthetized dogs. Vet Anaesth Analg 44:1313–1320
Sameem B, Saeedi M, Mahdavi M, Shafiee A (2017) A review on tacrine-based scaffolds as multi-
target drugs (MTDLs) for Alzheimer’s disease. Eur J Med Chem 128:332–345
Sánchez-Santed F, Colomina MT, Hernández EH (2016) Organophosphate pesticide exposure and
neurodegeneration. Cortex 74:417–426
Scarr E, Seo MS, Aumann TD, Chana G, Everall IP, Dean B (2016) The distribution of muscarinic
M1 receptors in the human hippocampus. J Chem Neuroanat 77:187–192
Schaal C, Chellappan S (2016) Nicotine-mediated regulation of nicotinic acetylcholine receptors in
non-small cell lung adenocarcinoma by E2F1 and STAT1 transcription factors. PLoS One
11:1–23
Sear JW (2019) Antihypertensive drugs and vasodilators. In: Hemmings HC, Egan TD (eds)
Pharmacology and physiology for anesthesia. Elsevier, Amsterdam, pp 535–555
Sehgal SA, Hammad MA, Tahir RA, Akram HN, Ahmad F (2018) Current therapeutic molecules
and targets in neurodegenerative diseases based on in silico drug design. Curr Neuropharmacol
16:649–663
Shahab L, Brose LS, West R (2013) Novel delivery Systems for Nicotine Replacement Therapy as
an aid to smoking cessation and for harm reduction: rationale, and evidence for advantages over
existing systems. CNS Drugs 27:1007–1019
Sherwood N (1993) Nicotine and psychomotor performance. Hum Psychopharmacol 8:155–184
Silver AA, Shytle RD, Sheehan KH, Sheehan DV, Ramos A, Sanberg PR (2001) Multicenter,
double-blind, placebo-controlled study of mecamylamine monotherapy for Tourette’s disorder.
J Am Acad Child Adolesc Psychiatry 40:1103–1110
Sivaraman D, Anbu N, Kabilan N, Kumar MP, Shanmugapriya P, Christian GJ (2019) Review on
current treatment strategy in ALZHEIMER’S disease and role of herbs in treating neurological
disorders. Int J Trans Res Ind Med 1:33–43
Smetana KS, Roe NA, Doepker BA, Jones GM (2017) Review of continuous infusion neuromus-
cular blocking agents in the adult intensive care unit. Crit Care Nurs Q 40:323–343
Svorc P (2018) Introductory chapter: autonomic nervous system - what we know about it. In: Svorc
P (ed) Autonomic nervous system. IntechOpen, London, pp 3–12
Takai K, Enomoto T (2018) Discovery and development of muscarinic acetylcholine M4 activators
as promising therapeutic agents for CNS diseases. Chem Pharm Bull 66:37–44
Takechi K, Suemaru K, Kiyoi T, Tanaka A, Araki H (2016) The α4β2 nicotinic acetylcholine
receptor modulates autism-like behavioral and motor abnormalities in pentylenetetrazol-kindled
mice. Eur J Pharmacol 775:57–66
Tangsucharit P, Takatori S, Zamami Y, Goda M, Poungrat Pakdeechote HK, Takayama F (2016)
Muscarinic acetylcholine receptor M1 and M3 subtypes mediate acetylcholine- induced
endothelium-independent vasodilatation in rat mesenteric arteries. J Pharmacol Sci 130:24–32
Taylor P (2012) Nicotinic receptors. In: Robertson D, Biaggioni I, Burnstock G, Low PA, Paton
JFR (eds) Primer on the autonomic nervous system. Academic Press, Cambridge, pp 79–82
Terry AV, Callahan PM (2018) Nicotinic acetylcholine receptor ligands, cognitive function, and
preclinical approaches to drug discovery. Nicotine Tob Res 21:383–394
Thakurathi N, Vincenzi B, Henderson DC (2013) Assessing the prospect of donepezil in improving
cognitive impairment in patients with schizophrenia. Expert Opin Investig Drugs 22:259–265
Thapa S, Lv M, Xu H (2017) Acetylcholinesterase: a primary target for drugs and insecticides. Mini
Rev Med Chem 17:1665–1676
104 S. F. Kokaz et al.
Zhao P, Sun H-X, Chu M, Hou Y-P (2016) Inhibitory effects of Botulinum toxin type a on pyloric
cholinergic muscle contractility of rat. Chin J Physiol 59:218–224
Zhao L, Chen M, Qian Y, Yang Q, Ge Y, Chen H, Qiu Y (2019) M1 muscarinic receptor activation
rescues β-amyloid-induced cognitive impairment through AMPA receptor GluA1 subunit.
Neuroscience 408:239–247. https://doi.org/10.1016/j.neuroscience.2019.04.007
Zheng G, Crooks PA (2015) Synthesis of lobeline, lobelane and their analogues. A review. Org
Prep Proced Int 47:317–337
Pharmacology of Adrenaline,
Noradrenaline, and Their Receptors 4
Bapi Gorain, Sulagna Dutta, Utpal Nandy, Pallav Sengupta,
and Hira Choudhury
Abstract
B. Gorain (*)
School of Pharmacy, Faculty of Health and Medical Science, Taylor’s University, Subang Jaya,
Selangor, Malaysia
S. Dutta
Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University,
Kuala Lumpur, Malaysia
U. Nandy
PK-PD, Toxicology and Formulation Division, CSIR-Indian Institute of Integrative Medicine,
Jammu, India
P. Sengupta
Department of Physiology, Faculty of Medicine, Bioscience and Nursing, MAHSA University,
Kuala Lumpur, Malaysia
H. Choudhury
School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
Keywords
Abbreviations
4.1 Introduction
Adrenaline and noradrenaline are two important catecholamines, which are respon-
sible for foremost activities in the maintenance of the “inner world” of the brain
body. The existence of these substances in the adrenal gland, which turns red upon
oxidation, was first discovered by Vulpian (Gaddum and Holzbauer 1957); however,
the pressor effect of the adrenal extracts was first recognized by Oliver and Schafer
(Oliver and Schäfer 1895). Later, adrenaline was isolated and within a few years,
adrenaline was synthesized chemically (Szymonowicz 1896). Later, the component
was known by two different names due to progress in research in two different
countries where Takamine referred to it as “adrenalin” (Takamine 1902) and “epi-
nephrin” was preferred by Abel (1899). Later, adrenaline is considered as its official
name in Britain, whereas in the United States, epinephrine is preferred.
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 109
Simultaneously, Addison revealed the necessity of the adrenal gland for life, and
in continuation to that some of the researchers thought cortisol from adrenal medulla
is the necessary principle responsible for the pressor effect (Addison 1855; Daven-
port 1982). Following researches over decades, noradrenaline is recognized as the
chief neurotransmitter from the postganglionic sympathetic nerve fibers (Gaddum
and Holzbauer 1957). Noradrenaline is also recognized as norepinephrine (similar to
adrenaline), which is the chief sympathetic neurotransmitter. It is known to control
tonic and reflexive changes in cardiovascular tone. Alternatively, adrenaline is a key
factor for the metabolic responses or for global challenges to homeostasis, including
expression of emotional distress. This adrenomedullary hormone is also involved in
preserving homeostasis in emergencies (Goldstein 2010).
4.1.1 Noradrenaline
4.1.1.1 Synthesis
Tyrosine, the circulatory neutral aromatic amino acid, is obtained biologically from
the hepatic hydroxylation of phenylalanine or from diet. This amino acid, upon
uptake into the para-aortic enterochromaffin cells of adrenomedullary cells, sympa-
thetic neurons, and specific centers in the brain, begins in synthesis of
catecholamines (Fig. 4.1). Tyrosine is then converted to L-dihydroxyphenylalanine
(L-DOPA) by the catalytic effect of tyrosine hydroxylase, where ionized iron,
tetrahydrobiopterin, and molecular oxygen potentiate this rate-limiting step of cate-
cholamine synthesis (Wassall et al. 2009). Later this L-DOPA is rapidly converted to
dopamine in the cytoplasm of neuronal cells by the help of DOPA decarboxylase
(L-aromatic-amino-acid decarboxylase), found mainly in the brain, liver, gut, and
kidney, in the presence of the cofactor pyridoxal phosphate. Finally, noradrenaline is
synthesized from dopamine by the catalytic action of dopamine-beta-hydroxylase,
the enzyme mostly confined to the noradrenergic cell vesicles, in the presence of
copper and ascorbic acid cofactor (Wassall et al. 2009). During increased
adrenomedullary and sympathetic outflow, may be due to stressor effect, the rate
of synthesis and presence of tyrosine hydroxylase increase simultaneously (Daubner
et al. 2011).
4.1.1.2 Metabolism
Noradrenaline is inactivated by taking up by the nerve cells, followed by storage of
the neurotransmitter or consequent intracellular metabolism. In this process, reup-
take by the noradrenaline transporters (uptake-1) on the nerve terminal cell mem-
brane plays the major pathway for terminating the action of noradrenaline
(Trendelenburg 1991). This uptake-1 process is carrier mediated and thus requires
energy to fulfill this action. Binding of adrenaline to the uptake-1 transporter does
not require catechol backbone; only one phenolic hydroxyl group containing drugs
are substrate to that site. Furthermore, the transport efficiency of the uptake-1
transporter decreases with increase in alkylation of the primary amino group of the
neurotransmitter. For example, reuptake efficiency of noradrenaline, adrenaline, and
isoprenaline (Fig. 4.2) is different, where reuptake of noradrenaline is highest
followed by adrenaline and isoprenaline (the extensively alkylated catechol)
(Burgen and Iversen 1965; Graefe and Bönisch 1988; Trendelenburg 1991).
As mentioned earlier, the uptake of noradrenaline via cell membrane mediated to
the axoplasm may undergo two fates, deamination to degrade by the action of
monoamine oxidases (Fig. 4.1) or may translocate into storage vesicles (Burgen
and Iversen 1965).
Thus, vesicular storage and degradation of the axoplasmic noradrenaline result in
an intra-neuronal “sink” with very low concentration of noradrenaline in cytoplasm.
The metabolism of catecholamines through oxidative deamination by the mono-
amine oxidases is common in neural and non-neural tissues. Rapid uptake of the
axoplasmic noradrenaline in the vesicles maintains the concentration of monoamine
oxidases onto the surface of mitochondria for continuous functioning of catechol-
aminergic systems (Goldstein 2010). Anything that prevents these monoamine
oxidases can increase the cytoplasmic concentration of catecholamines, stimulate
the outward transport of noradrenaline, and thereby promote cardiac smooth
muscles, vasoconstriction, leading to hypertension.
These monoamine oxidase enzymes are the chief intracellular enzyme that is
involved in metabolizing xenobiotic or biogenic amines throughout the biological
system (Westlund et al. 1988). Over a half of a century ago, Johnston brought two
4
Tyrosine
OH
3,4-dihydroxy phenylglycol
L-DOPA
CHO COMT
Aldehyde reductase
α2 OH
Noradrenaline COMT
3-methoxy-4hydroxyl phenylglycol
OH
MAO O
CHO
Noradrenaline 3,4-dihydroxy mandelic acid
OH
Aldehyde dehydrogenase
α2
Gq Gs
Gi
Fig. 4.2 Chemical structure of (a) noradrenaline, (b) adrenaline, (c) isoprenaline
111
112 B. Gorain et al.
4.1.2 Adrenaline
4.1.2.1 Synthesis
Adrenaline synthesis follows the similar synthetic pathway from tyrosine, as
described in the synthesis process of noradrenaline. Following the synthesis of
noradrenaline, it is converted to adrenaline by the catalytic action of
phenylethanolamine-N-methyltransferase, where adrenocortical steroidal hormones
(e.g., cortisol) act as trophic for this catalytic enzyme (Hillarp and Hokfelt 1955;
Wurtman and Axelrod 1966).
It has been postulated that the uptake-1 in sympathetic nerve fibers usually takes
up noradrenaline along with the circulatory adrenaline. Upon stimulation of the
sympathetic fibers, the nerve terminals release noradrenaline along with the up taken
adrenaline. This coreleased adrenaline binds to specific β-adrenoceptors on the nerve
terminal, subsequently increasing the release of noradrenaline from the terminal
(Iversen 1971). This hypothetical phenomenon brings up a model where the endog-
enous components are taken up into the nerve terminals, coreleased with the
neurotransmitter, and finally exaggerate or prolong the release pattern through
binding at facilitatory presynaptic receptors. However, confirmation of this hypo-
thetical assumption in isolated tissue preparation in in vitro models has failed
(Goldstein 2010).
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 113
4.1.2.2 Metabolism
The presence of catechol-O-methyltransferase in the medullary cells of adrenal
cortex leads to the formation of adrenaline from noradrenaline, simultaneously
resulting in inactivation of adrenaline following a similar pathway of noradrenaline.
As a result of metabolism, catechol-O-methyltransferase produces metanephrine,
thereby decreasing cytosolic concentration of adrenaline. Thus, the production of it
is continuous in the cytosol, whereas the release of adrenaline from the
adrenomedullary vesicles is episodic, upon receiving the impulse. Pheochromocy-
toma is a type of chromaffin cell tumors, where synthesis of catecholamines
increases heavily and thus, increases the metanephrines in the body. Therefore, the
detection of the level of metanephrines in plasma provides information on increased
levels of catecholamines, to aid effective treatment of the disease (Lenders et al.
2002; Spence et al. 2018).
Studies in the last few decades enabled developing concepts regarding different
receptors that mediate actions owing to the binding of endogenous catecholamines.
These were previously divided into only α- and β-adrenoceptors or ARs, while later
in the 1970s the α adrenoceptors are further divided into α1 and α2 ARs. The α1 ARs
are generally designated to mediate the responses in the effector organ. The α2 ARs
are generally localized in the presynaptic knob to regulate the release of
neurotransmitters, while reports also suggest their postsynaptic locations. Both α1
and α2 ARs are important in regulating the vascular tone, while none are known to
include homogenous groups. The initial division of the α1- and the α2-AR subtypes
114 B. Gorain et al.
was purely based upon the pharmacological properties of the same, but later detailed
structural analyses were performed by isolation and identification via cloning
methods. Today, α1-ARs have been classified into α1A-AR subtype (formerly
α1a/c, cloned α1c), α1B-AR subtype (cloned α1b), and α1D-AR subtype
(formerlyα1a/d, cloned α1d). It may convincingly be suggested that the α1A-AR
subtype is more prominently designated for vascular basal tone and arterial blood
pressure (BP) regulation, while α1B-AR subtype is better implicated for responses
towards exogenous agonists. The α1B-AR subtype expressions may also get altered
or modified owing to various pathophysiological modifications. The α2-AR
classifications include α2A/D-AR subtype, α2B-AR (cloned as α2b), and α2C-AR
(cloned as α2c). The α2A and α2B ARs are mainly used to mediate arterial
contraction, while α2C-ARs are mainly designated for venous vasoconstriction.
The functions of the α-ARs are subtype specific as well as common, depending
upon the required responses of the effector tissue. In most of the cases, the responses
evoked different α‐AR subtypes do overlap due to the lack of receptor specificity of
some ligands. Thus, it is essential to design drugs that will be specific in their mode
of actions in vivo, for therapeutic intervention using these receptors.
4.2.1.1 a1-Adrenoceptors
Structural Biology
α1-ARs mediate the essential role to regulate the physiological action of norepineph-
rine and epinephrine. The α1A, α1B, and α1D represent the three α1-AR subtypes.
They belong to the GPCR family and act via the Gq/11 signaling pathway. They
display individualistic patterns of tissue distributions and pharmacological
properties (Cotecchia 2010).
The pioneer hypothesis of Easson-Stedman had initiated bulk of research directed
towards drug designing for the ARs. Once ARs were cloned and the subtypes had
surfaced, the studies on drug design had oriented to reveal the detailed structure of
the receptors and new interactions were found. The α1 subtype and β-ARs were
found to greatly differ in their ligand-binding pocket which accounted for selective
drug design (Easson and Stedman 1933). Ahlquist, in later years, had used a series of
agonists to reveal the AR subtypes as α and β, and further into α1 and α2 receptors as
post- and pre-junctional ARs respectively (Starke et al. 1989; Ahlquist 1948). The
development of advanced pharmacological techniques, such as the radioligand
binding assay, leads to the classifications of α1-ARs into α1A and α1B-subtypes
(Morrow and Creese 1986). This was followed by the interventions of Art Hancock
who revealed the structure-function association of α1-ARs (Hancock et al. 1988) and
led to the synthesis of the first synthesized selective agonist α1A-AR, A-61603
(Knepper et al. 1995).
Initially four subtypes of α1-AR were revealed by cloning techniques. The α1b-
AR subtype was the first cloned α1-AR subtype, which displayed similar binding
properties like that of α1B-AR. Besides this, there were α1a from rats (Cotecchia
et al. 1988), bovine α1c- (Schwinn et al. 1990) and rat α1d-AR (Perez et al. 1991),
which had 99.8% homogeneity and considered a same subtype. At present, it is
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 115
conceived that the α1a/α1d clones refer to a novel α1D subtype. The latest classifi-
cation includes α1A (former α1a/c), α1B (former α1b), and α1D (former α1a/d)
(Perez et al. 1994).
Biological Distribution
The tissue distribution for α1-AR varies with their subtypes and depends on the
specific functions. Experiments in human cells, rats, and rabbits have showed that
the α1A/c-ARs are highly expressed in the heart, liver, salivary gland, lung, and vas
deferens (Table 4.1) (Schwinn et al. 1990; Perez et al. 1994). The mRNA expression
and receptor protein expressions also revealed the expression of this receptor
subtype in rat kidney, mediating vasoconstriction. In situ hybridization analysis of
α1-AR-mRNAs in rat brain displayed substantial levels of α1A-ARs expressions in
the olfactory system, hypothalamus, brainstem, and spinal cord, especially in the
structures that play roles in motor functions. The expression of α1A-ARs has also
been reported in glial cells, thus suggesting their non-neuronal mode of action in
mediating several brain functions (Day et al. 1997).
The α1B-AR has been reported to be highly expressed in rat liver and heart while
less in the hippocampus, aorta, and salivary gland. The expression of this receptor
subtype is high in the pineal gland, the lateral nucleus of the amygdala, most of the
thalamic nuclei, and in the raphe nuclei (Day et al. 1997).
The α1D-AR has been found to be abundant in the deferens and aorta and less
expressed or absent in the liver, spleen, kidney, and salivary gland. In neural tissues,
the distribution of this subtype is the most unique. It is highly expressed in the
olfactory system, hippocampus, cortical layers II–V, amygdala, the reticular
thalamic nucleus, the motor nuclei of the brainstem, and in the spinal cord (Day
et al. 1997).
Subtype-Specific Functions
Functions Mediated by a1A-ARs
The α1A-ARs have been suggested to play a vital role in mediating contractions in
several tissues such as in the vas deferens (Moriyama et al. 1997; Burt et al. 1998),
renal artery (Villalobos-Molina et al. 1997), rabbit ear artery (Fagura et al. 1997), tail
artery in rats (Lachnit et al. 1997), right atrium (Yu and Han 1994), internal anal
sphincter (Mills et al. 2008), and in the prostate (Marshall et al. 1995). α1A-ARs in
the rat vas deferens display two responses. One of these responses is phasic, may be
due to calcium ion release from ryanodine-sensitive compartments, and the other one
is tonic response which may be mediated via the pathway involving protein
kinase C, diacylglycerol, and calcium ion influx through L-type and may be also
the T-type channels (Burt et al. 1998). The rat submandibular gland may contain
116
Table 4.1 Classifications, functions, and phenotypes of α1-adrenoreceptor subtype in mice with genetic modifications
AR Previous
group Present pharmacology Cloning Functional responses Genetic modification Phenotype
α1Ha α1A α1A α1c Control of blood pressure; Overexpression/ Increased contractile response, no
(α1A/ vasoconstriction; smooth muscle heart-specific hypertrophy
c) contraction promoter
α1B α1B α1b Regulatory; minor contractile role Gene deletion Decreased resting blood pressure,
decreased vasoconstriction
α1D α1D α1a, Control of blood pressure; Gene deletion Decreased resting blood pressure,
α1d, vasoconstriction; smooth muscle decreased vasoconstriction
α1a/d contraction
α1Lb α1Nc α1N –
α1L α1L – Vasoconstriction
a
High affinity for prazosin vs α1L-AR group
b
Members of this group have not yet been cloned
c
High affinity for HV 732 vs α1L receptors. Poorly classified with available probes
B. Gorain et al.
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 117
both α1A- and α1B-ARs, but in several experimental setup, it serves as a model for
the α1A-AR ligand-binding sites (Bruchas et al. 2008). It has been observed that
positive inotropic effects of phenylephrine in mouse are mediated via its binding to
the α1A-ARs (Ross et al. 2003). Moreover, noradrenaline-mediated contractions in
the prostate are also evident to occur via the α1A-AR (Gray et al. 2008). It is
suggested that overexpression of the α1A-AR elevates β-AR-supported cardiac
muscle contractility, thereby ameliorating the outcome from myocardiac infarction
(Woodcock 2007).
Physiological Functions
Control of Blood Pressure
α1-ARs in the vascular system mainly mediate contractions and contribute greatly in
blood pressure regulation and in the baroreceptor reflex response. Piascik et al.
(1990) had put forth that in the conscious rat, α1A-AR subtype mediates tonic
maintenance of blood pressure, while α1B-AR subtype mediates responses towards
the exogenous agonists. In the unconscious or pithed rat, it has been reported that
both the blood pressure reflex and actions of exogenous noradrenaline involve both
α1A- and α1D-ARs, while blood pressure regulation via noradrenaline is mostly α1-
AR mediated (Vargas and Gorman 1995).
Temperature Control
Temperature regulation is another essential function of vascular α1-ARs since
vasoconstriction of superficial blood vessels is the primary mechanism for the core
body heat conservation. Methylenedioxymethamphetamine (MDMA) is an exten-
sively used recreational drug, which may lead to life-threatening hyperthermia. In
animal experimentations, MDMA reportedly affects thermoregulation, often leading
to hypothermia or hyperthermia in cold or hot ambient temperatures, respectively.
The α1-AR (α1A/D-ARs) antagonists could potentially covert the MDMA-mediated
monophasic hyperthermic response to a biphasic response characterized by hypo-
thermia followed by hyperthermia (Docherty 2010).
Depression
Depression, a multifactorial disorder, involves innumerable neural circuits and
neuronal processes. Noradrenaline plays significant roles in depression via the
ARs (Morilak et al. 2005). The ARs were found to undergo alterations followed
by the administration of antidepressant drugs (Blier 2003). Brain α1-adrenergic
neurotransmission impairment has been reported in certain depressive illnesses. It
is not yet clearly understood what is the exact mechanism by which individual α1-
AR subtypes mediate antidepressant effects. It had been suggested that α1-AR is
specifically associated with antidepressant action via a study using chronic
treatments with imipramine and electroconvulsive shock. The observations revealed
an increase in α1A-AR and not in the α1B-AR mRNA land receptor expressions in
the rat hippocampus and cerebral cortex (Stone and Quartermain 1999).
Nociception
The noradrenergic system is well known for its analgesic effects (Jinushi et al. 2018).
A majority of the studies that are focused on noradrenaline-mediated analgesic
effects have reported the antinociceptive role of α2-AR, while studies on the role
of α1-AR are mostly behavioral ones (Wei et al. 2016; Di Cesare et al. 2017). The
α1-AR binding modulations due to pain mostly take place in the areas of the central
nervous system (CNS) involved in pain processing. The receptor modulations,
including those of the α1B-AR subtype in acute pain phase, have been suggested
to be often lateralized and to vary according to the pain phases, whereas α1-ARs
involved in the late phases of pain were other than the α1B. Research on α1D-AR-
deficient mice [19] that focused on responses to different noxious stimuli put forth
the concept that α1D-ARs in the spinal cord participate in mediating responses of
thermal pronociception (Dogrul et al. 2006).
120 B. Gorain et al.
Genitourinary Functions
Three α1 AR subtypes have been found to be localized in the lower urinary tract:
α1A, α1B, and α1D. α1A receptors are predominant (about 70%) in the stroma of the
prostate gland, α1B in the prostate gland epithelium, and α1D mostly in the prostate
blood vessels as well as in the stroma. The α1ARs are also located in the urinary
bladder and urethral smooth muscle, as well as in the spinal cord and ganglia. The
extraprostatic sites of α-receptors lead to reduced organ selectivity and cause side
effects of treatments that focus on modulations of these receptors for lower urinary
tract symptoms. The α-blockers are used to treat benign prostatic hyperplasia (BPH)
owing to direct α-adrenergic antagonism of smooth muscle tonicity in prostatic
stroma. Current research has revealed longer-term effects of α-blockers on prostate
cellular differentiation and apoptosis (Cavallo 2018).
Receptor Modulators
Selective α1-adrenergic modulators retain a tiny low market share among all antihy-
pertensive drug medications within the United States (Griffith 2003). Prazosin,
introduced in 1976, was the primary marketed drug in this category. Afterwards,
two more α1-adrenergic blockers, doxazosin and antihypertensive drug, were avail-
able on the market, and their potential for once-daily dosing has provided extra
treatment flexibility (Xie et al. 2005). Within the last decade, sustained release
formulations for alpha-blocker and doxazosin are revealed. Tamsulosin and
alfuzosin are so-called uroselective agents with a better affinity for prostate α1-
ARs and are unremarkably employed in the management of patients with BPH
(White and Moon 2005).
α1-adrenergic antagonists, both selective and nonselective, are clinically avail-
able. Phenoxybenzamine, a nonselective, noncompetitive blocker, is currently
reserved for the preoperative treatment of pheochromocytoma-associated high
blood pressure. Nonselective α-blockade means that presynaptic α2-receptors,
which cut back the discharge of noradrenaline, are downregulated because of
inhibited feedback mechanism. Phentolamine could be a short-acting, competitive,
nonselective α-blocker parenterally administered and used for severe sorts of high
blood pressure prompted by extreme release of catecholamines. Prazosin, the pio-
neer selective α blocker, contains high affinity for the α1-AR associated with an
immediate-release formulation and has speedy onset of action. These features most
likely account for its comparatively higher rate of syncope and postural hypotension
compared with terazosin and doxazosin. Syncopal episodes may be decreased by
limiting the initial dose to 1 mg, administering the primary dose at night before sleep,
and gradually increasing the dosage. Doxazosin and terazosin have lower lipid
solubility than prazosin and thus bear lesser affinity for α1 receptors.
Different α1-adrenergic antagonists are pharmacologically discrete. Prazosin
features a comparatively short period of action and should be provided in a minimum
of double doses daily (Stanaszek et al. 1983). Doxazosin and terazosin with longer
half-lives might be given once daily. Doxazosin should be administered at bedtime,
which appears to be safe and effective in reducing morning hypertension (Pickering
et al. 1994; Fulton et al. 1995). α1-adrenergic antagonists ought to be used
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 121
cautiously in pregnant women and in children, since the effectualness and/or safety
of those compounds have not been estimated.
α1-adrenergic-specific antagonists hinder the noradrenaline-mediated vasocon-
striction via selective inhibition of postsynaptic α1 receptors activation by
catecholamines (Lund-Johansen and Omvik 1991). The presynaptic α2-ARs are
unblocked with these selective compounds, so inhibition of vasoconstriction by a
feedback mechanism of α2-AR stimulation is preserved.
The norepinephrine inhibition may be the cause of the absence of tachycardia,
higher cardiac output, and increased renin levels by the antagonists of both presyn-
aptic α2 ARs and the postsynaptic α1 ARs such as phentolamine (Baez et al. 1986).
As these drugs do not affect the renin-angiotensin-aldosterone system, they are
suited to be employed for hypertension regulation in patients with disorders related
to this axis (Webb et al. 1987). These drugs have ameliorative effects on
hemorheology as well including blood cell disorders, blood viscosity, and endothe-
lial function (Gomi et al. 1997).
The α1-AR antagonists may cause orthostatic hypotension in patients with either
volume-depletion disorders or susceptible to the loss of α1-ARs-induced vasocon-
striction. The actions of the α1-AR antagonists are at per with the sympathetic
activation, which is the reason for unchanged blood pressure after administration
of α1-AR antagonists in normotensive persons with normal activities of sympathetic
nervous system.
Structural Biology
As discussed earlier, ARs were first classified as α- and β-receptors by Raymond
Ahlquist in 1948 on the basis of their pharmacological actions in various tissues
(Ahlquist 1948). With the advent of research in this realm, the adrenergic or
“adrenotropic” receptors were divided into three primary classes and into nine different
mammalian subtypes (Ahlquist 1948). The recent classification of receptors into three
main classes, α1-, α2-, and β-ARs, mainly relies on their amino acid sequences along
with pharmacological characteristics. Each of these classes of the ARs has again been
sequestered into three subtypes (Hieble et al. 1995). For α2-adrenoceptor, three subtypes
were previously referred to as α2-C10 (Kobilka et al. 1987), α2-C2 (Lomasney et al.
1990), and α2-C4 (Regan et al. 1988) according to their gene locations on human
chromosomes. These are now better recognized as AR subtypes α2A, α2B, and α2C.
α2-ARs are membrane glycoproteins whose most structural features bear resem-
blance and are common to other GPCRs. The common feature includes the presence
of seven transmembrane (TM) domains with extracellular amino terminus and
intracellular carboxyl terminus. The receptor binding sites must be accessible to
the specific ligands like adrenaline and noradrenaline. Thus, the binding sites of α2-
122 B. Gorain et al.
ARs are positioned within the core of the receptor proteins comprising the seven
α-helical TM domains (Laurila 2011).
The unique structural characteristics have rendered the α2-ARs-ligand
interactions to be highly specific. The ligand binding cavity in these receptors has
been reported to be structured by the residues in the third, fifth, sixth, and seventh
TM as well as the second extracellular loop. The relative lower affinity of dopamine
at α2-ARs than that of noradrenaline may be due to the absence of a specialized
β-hydroxyl moiety in dopamine molecule (Nyrönen et al. 2001). Besides these, the
other residues those supposedly have hydrophobic interactions with the N-methyl
group (positively charged) of catecholamine ligands are two phenylalanines posi-
tioned at 7.38 and 7.39 in the seventh TM. In the fifth TM of the human α2A-
adrenoceptor, two serine residues expose sites for hydrogen bonding to two
catecholic hydroxyl groups of the catecholamine ligands. These interactions may
be altered by conformational changes through receptor activation. They are also vital
for ligand orientations within the ligand-binding site of the α2-ARs. Some residues
mediate π-π stacking interactions with the aromatic ring present in the
phenylethylamine-type ligands (Nyrönen et al. 2001).
Unlike the agonists of α2-ARs, the concepts regarding receptor structural
characteristics of antagonists binding for these receptors are less clear. The α2-AR
antagonists bear way higher chemical diversity than that of agonists. Thus, the
antagonists are much complex and possess more divergent modes of binding.
There is no report that could specify the “antagonist binding site” for adrenoceptors
or for other GPCRs. However, in α2-ARs, a monoamine-binding GPCR, antagonists
have been predicted to bind almost at the same orthosteric agonist-binding sites
(Nyrönen et al. 2001).
Biological Distribution
The distributions of α2-AR subtypes in the mammalian tissues, especially in humans
and in rodents, have been detected using various techniques. Receptor radioligand
binding assays and autoradiography were employed to detect ligand binding
(Boyajian and Leslie 1987); in situ hybridization and other mRNA quantification
methods were used to determine receptor gene expression (Nicholas et al. 1993), and
antibody-based techniques such as immunohistochemistry and Western blotting
were used for receptor characterization (Tran et al. 2004). The three α2-AR subtypes
have exclusive tissue distribution patterns in the CNS, peripheral nervous system,
and in the peripheral tissues. The expressions of α2A-AR have been found exten-
sively in peripheral tissues and in the CNS, i.e., brainstem (especially the locus
coeruleus), midbrain, hypothalamus, hippocampus, spinal cord, cerebral cortex,
cerebellum, and septum. The expressions of α2B-adrenoceptor have been observed
mostly in the peripheral tissues and in lower levels in the CNS, mainly in the
thalamus, olfactory system, pyramidal layer of the hippocampus, and cerebellar
Purkinje layer. The α2C-ARs seem to be expressed mostly in the CNS, with different
expression patterns from those of α2A-ARs. They are most abundant in the cerebral
cortex, midbrain, amygdala, thalamus, olfactory system, dorsal root ganglia, hippo-
campus, basal ganglia substantia nigra, striatum, and ventral tegmentum (Brodde
et al. 2001) (Table 4.3).
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 123
Table 4.3 Tissue distributions of α2-adrenoceptor subtypes in the central nervous system and
peripheral tissues with their functions
Receptor Peripheral
subtype CNS tissues Physiological functions
α2A Amygdala Kidney Analgesia
Locus Vasculature Bradykinesia and hypotension
coeruleus
Lateral Urethra Hypothermia
septum
Brainstem Heart Inhibition of epileptic seizures
Cerebral Platelets Presynaptic inhibition of neurotransmitter
cortex release
Thalamus Spleen Anxiety like behavior
Hypothalamus Salivary Sedation and anesthesia
glands
Hippocampus Pancreas Regulation and blood glucose and insulin
Spinal cord Fat cells Decrease in intraocular pressure
Olfactory Inhibition of gastrointestinal motility
nucleus
Retina
α2B Thalamus Kidney Placental angiogenesis
Placenta Salt-induced hypertension
Liver Vascular smooth muscle contraction
Vasculature
α2C Striatum Kidney Presynaptic inhibition of catecholamine
release
Olfactory Adrenal Modulation of motor behavior
tubercle gland
Locus Vasculature Regulation of dopamine and serotonin
coeruleus balance in the brain
Hippocampus Pancreas Vascular smooth muscle contraction
Cerebral
cortex
Amygdala
Substantia
nigra
reduction in intracellular cAMP level and thereby protein kinase A (PKA) remains
inactivated. The PKA-mediated phosphorylation of downstream proteins such as
phosphorylase kinase, an enzyme for glycogen metabolism, is thus inhibited (Starke
et al. 1989).
The α2-ARs have slightly higher affinity in binding noradrenalin released from
the sympathetic postganglionic fibers than adrenaline from the adrenal medulla
(Boron and Boulpaep 2003). These receptors mediate several common functions
as the α1-AR, while having few specific physiological roles as well, including
sympathetic inhibition analgesia, sedation, hypotension, increased opioid and alco-
hol withdrawal symptoms, pupil control, body temperature regulation, seizure sus-
ceptibility modulation, and blood glucose homeostasis. The α2-AR agonists are
often used as veterinary anesthetics owing to its functions to cause analgesia,
sedation, and muscle relaxation via acting upon the CNS (Khan et al. 1999).
Presynaptic Regulation
In central adrenergic nerves as well as in sympathetic nerves, α2A- and α2C-ARs
regulate the neurotransmitter release by acting as inhibitory autoreceptors. α2B-
receptors on postsynaptic cells are activated by catecholamines released via the
sympathetic nerves. By doing so, they mediate various physiological functions
such as vasoconstriction. Functional differentiation of the presynaptic α2A- and
α2C-ARs has been proposed such that α2A-ARs have been found to inhibit the
release of norepinephrine from sympathetic nerves initially at high stimulation
frequencies, while the operational frequency for the α2C receptor can be very low
in order to regulate basal norepinephrine release (Philipp et al. 2002).
Analgesia
All the three α2-AR subtypes play vital roles in the regulation of pain perception
(Bücheler et al. 2002). α2-ARs present in high levels in the spinal cord, specifically
in the superficial layers of the dorsal horns, are reported to regulate incoming
nociceptive impulses. α2A-ARs are essential to mediate the analgesic effect of α2-
AR agonists, the spinal α2C-ARs are reported to effectuate the moxonidine-
mediated analgesia, while the α2B-ARs are needed for spinal antinociception caused
by nitrous oxide (Philipp et al. 2002).
Sedation
α2-ARs agonists serve as potent sedatives and hypnotic agents in intensive care as
well as in the postoperative phase. The hypnotic effects mediated by α2-ARs are
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 125
suggested to be regulated by the locus ceruleus, since its neurons express high levels
of α2A-ARs (Philipp et al. 2002; Nguyen et al. 2017).
Behavior
α2-ARs mediate several behavioral functions owing to their high levels of
expressions in multiple sites in the CNS (Schramm et al. 2001; Frances Davies
et al. 2004). They have been shown to impede sensory information processing by the
CNS. They may also cause locomotor inhibition. α2A- and α2C-ARs have comple-
mentary actions in the integration of CNS function and behavior (Björklund et al.
2001; Philipp et al. 2002).
Receptor Modulators
α2-blockers represent the alpha blocker subset of drugs that are antagonists to the
α2-ARs (Rang et al. 1999). These drugs are mostly used in research arena, still not
widely applied in clinical interventions for humans. Their functions are based on
blockade of α2-ARs and thereby increase in the release of noradrenaline. These
α2-AR antagonists have been shown to significantly increase the release of dopami-
nergic and serotonergic besides the adrenergic neurotransmitters, trigger insulin
secretion, and reduce blood sugar levels (Rang et al. 1999).
Yohimbine is the most commonly used antagonist of α2-ARs (Rang et al. 1999).
It still finds immense applications in veterinary medicine, alongside its potent
alternative, atipamezole (Haapalinna et al. 2003; Lemke 2004). They operate to
reverse the effects of α2-AR agonists (such as medetomidine) that are used as
sedatives during surgery (Lemke 2004). Other α2-AR antagonists are efaroxan,
idazoxan, phentolamine, and rauwolscine (Chopin et al. 1999).
α2-AR antagonists find implications in the treatment of depression. The
tetracyclic antidepressants mirtazapine and mianserin are two of the antidepressants
belonging to this class of antagonists, but their antidepressant effects are also
supported by activation of other receptor sites as well. Sudden withdrawal from
use of the α2-AR antagonist drugs can prove detrimental because an immediate
global downregulation of release of the neurotransmitters may lead to various
neurological problems, depressions and sudden hyperglycemia, and reduction in
insulin sensitivity triggering diabetes. Moreover, downregulation of microcircula-
tion as well as adrenaline hypersensitivity in organs like the liver may also result if
there is a sudden withdrawal of a regular α2-AR treatment (Rang et al. 1999).
126 B. Gorain et al.
Three different types of β adrenoceptors (β1, β2, and β3) are identified so far with a
homology of approximately 60% amino acid sequence. Ligands, such as adrenaline
and noradrenaline, bind to the ligand-binding pocket of the receptor site to control
various functional responses including contractility and heart rate, relaxation of smooth
muscle, and numerous metabolic events. As discussed earlier, all these receptor
subtypes are coupled to G-protein Gs and simultaneously activate adenylyl cyclase
(Table 4.4). However, research findings suggest variance in moderation of events and
signals, such as receptor downregulation or desensitization, following activation of
three different β-ARs (Lefkowitz 2000; Ma and Huang 2002; Kohout and Lefkowitz
2003). However, the magnitude of such regulation largely depends on the affinity of the
receptor to bind with the ligand, and thus, β2 ARs are found more susceptible. Overall,
the activation process of β-receptors leads to the accumulation of cyclic adenosine
monophosphate (cAMP) followed by activation of PKA and phosphorylation, thereby
alteration of function of cellular protein (Table 4.4).
4.2.2.1 b1-Adrenoceptor
Structural Biology
One of the major subtypes of the β-ARs is the β1 receptor (Bylund et al. 1994). The
receptors consist of seven membrane-spanning domains, three intra- and three extracel-
lular loops, one extracellular N-terminal domain, and one intracellular C-terminal tail.
Structural determination of β1 ARs was challenging because of its purification
difficulty and instability in detergent (Warne et al. 2008). However, β1 ARs from
frog and turkey red blood cells were estimated to have a molecular weight of 54,000
and 43,500, whereas β1 ARs obtained from mammalian lungs were shown to have a
molecular weight of 60,000–65,000 (Shorr et al. 1985). In fact, the first high
resolution picture of the ARs is obtained from β ARs, where the ligand binding
pockets were clear to fit a selective ligand (Suryanarayana et al. 1992).
Biological Distribution
β1-ARs are dominantly present in the cardiac tissue, coronary artery, adipose tissue,
and juxtaglomerular cells of kidney (Wallukat 2002). The presence of β1-AR has
also been found in the central nervous system, in brain (Gingrich and Caron 1993).
Table 4.4 Beta (β) adrenergic receptors and their effector system
Receptor subtype G protein Examples of some biochemical effectors
β1 Gs " adenylyl cyclase,
" L-type Ca2+ channels
β2 Gs " adenylyl cyclase,
β3 Gs " adenylyl cyclase,
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 127
Receptor Modulators
β1-AR agonists are used for cardiac stimulation in the treatment of cardiogenic shock
and severe congestive heart failure (CHF) (Wallukat 2002). Dobutamine,
denopamine, and xamoterol are the important members in this category. Though
dobutamine acts on both α and β-ARs, its specificity towards β1-AR is relatively
higher for which it has been considered as a selective β1-AR agonist (Parker et al.
2008). Denopamine is also a β1-AR agonist and is used in the treatment of angina
(Ishide 2002; Nakajima et al. 2006). It can also be used to treat CHF and pulmonary
edema (Nishio et al. 1998; Sakuma et al. 2001). In contrast, xamoterol is a partial
agonist to β-AR where it stimulates the heart at rest and blocks during exercise (Rang
et al. 1999). On the other hand, β-AR blockers are classified into β1-AR blocker
(cardioselective) and nonselective blocker, i.e., they block both β1 and β2-AR. In all
cases associated with β-AR blockers, ‘selectivity’ is a relative term and not absolute.
Therefore, selective β1-AR blocking may be associated with β2-AR-mediated
bronchoconstriction but less likely to produce these side effects (Baker et al. 2017).
Metoprolol, atenolol, acebutolol, bisoprolol, esmolol, betaxolol, celiprolol, etc. are the
more potent blockers of β1-AR than β2-AR and produce better cardioselective actions
depending upon the dose. These drugs are used to treat patients depending upon the
associated properties of their own viz. with intrinsic sympathomimetic action, without
sympathomimetic action, with additional α-AR blocking ability, membrane stabilizing
action etc. as well as patient’s viz. diabetes, asthmatics, etc. (Rang et al. 1999). On this
basis, there are numerous clinical uses of β-AR antagonists like hypertension, angina
pectoris, myocardial infractions, cardiac arrhythmias, glaucoma, etc. Metoprolol is the
classical molecule as a selective β1-AR blocker. It shows weak membrane stabilizing
activity but devoid of intrinsic sympathomimetic activity. Acebutolol is another
cardioselective drug with partial agonistic effects, i.e., it activates β1 and/or
β2 submaximally and membrane stabilizing actions. It is preferred for patients prone
to bradycardia. It has also intrinsic sympathomimetic activity. Diacetolol is also a
128 B. Gorain et al.
β-adrenergic blocker which is the primary metabolite of acebutolol and used as an anti-
arrhythmic agent (Basil and Jordan 1982). Atenolol and bisoprolol are selective β1-AR
blockers without any intrinsic sympathomimetic action and primarily used to treat high
blood pressure as well as heart-associated chest pain. Bisoprolol has a higher degree of
β1-AR selectivity compared to other selective β1-AR β blockers like atenolol, meto-
prolol, and betaxolol. However, nebivolol has better selectivity than bisoprolol.
Esmolol is an ultrashort acting β1-AR blocker and preferably used during cardiac
surgery to control heart rate and BP (Jaillon and Drici 1989; Deng et al. 2006).
Betaxolol is a selective β1 AR blocker, which is used for the treatment of hypertension
as well as glaucoma. Its therapy may be advantageous over timolol with respect to
fewer β2-AR blocking mediated side effects. The levo form of betaxolol is also
available on the market. Celiprolol is a selective β1-AR blocker with additional β2-
AR partial agonistic activity that is beneficial for asthmatic patients to avoid worsening
of disease conditions. It is also a weak α2 receptor antagonist. It has orphan drug status
for the treatment of vascular Ehlers–Danlos syndrome through promoting normal
collagen synthesis in the blood vessels and thereby shifting the pressure load away
from the vessels prone to rupture (Beridze and Frishman 2012). Flusoxolol is also a
selective β-AR blocker. Landiolol is a selective β1-AR antagonist that reduces heart
rate effectively with less negative effect on blood pressure or myocardial contractility
(Ikeshita et al. 2008; Wada et al. 2016). This is also an ultra short-acting drug like
esmolol but shows better cardioselectivity (Baker 2005; Okajima et al. 2015).
4.2.2.2 b2-Adrenoceptor
Structural Biology
β2 ARs are another key subtype of β-ARs which also belong to GPCRs superfamily
(Bylund et al. 1994). The representation of the binding pockets of β2 ARs from
humans is made in Fig. 4.3, where only one antagonistic binding site differs between
β1 ARs and β2 ARs constituting 15 amino acids. The presence of tyrosine in β2 AR
at the top of TM7 differs by the presence of phenylalanine in β1 AR (Suryanarayana
et al. 1992). The represented extracellular surface of β2 AR in Fig. 4.3b shows the
differences with β1 AR in yellow. The interior of the β2 AR has been represented in
Fig. 4.3c, where the binding pockets are split, with TM6-TM7 on the left and
TP-TM5 on the right (Kobilka 2011). The figure represents the identical nature
between the structures of β1 AR and β2 AR from the binding pocket to the
cytoplasm. Thus, it had been inferred that the structural diversity of the amino
acids are much higher into the binding pockets (Kobilka 2011).
Biological Distribution
β2-ARs are located mainly in the smooth muscles of the biological system, which
include vascular, airway, gastro-intestinal tract, uterus, bladder sphincter, seminal
tract, iris (radial muscle), and ciliary muscle (Bylund et al. 1994).
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 129
Fig. 4.3 Subtype-specific ligand binding in the β2AR. Amino acids that differ between β1AR and
β2AR are shown in yellow. The inverse agonist carazolol is shown with green carbons. (a) The
binding pocket of the human β2AR. Only one of the 15 amino acids that constitute the antagonist
binding pocket differs between β1AR and β2AR. Tyr 308 at the top of TM7 in the β2AR is Phe in
the β1AR. (b) The extracellular surface of the β2AR. (c) The interior surface of the β2AR that has
been split along the plane of the binding pocket (Kobilka 2011)
130 B. Gorain et al.
Receptor Modulators
Many selective β2-ARs agonists such as salbutamol, terbutaline, salmeterol, and
formoterol have been developed for their bronchodilator action for the relief of
reversible airway obstruction like asthma. These drugs have lower incidence of side
effects mediated by β1-ARs whereas long-term use of it led to muscle tremor through
change in β2-ARs-mediated rate and force of contraction. The activation of β2-ARs in
the α-cells of pancreatic islets causes increase in glucagon secretion. Isoxsuprine is a
selective β2 ARs agonist that causes direct relaxation of uterine and vascular smooth
muscle via β2-ARs. Isoxsuprine is used as a tocolytic for the treatment of premature
labor and as a vasodilator for the treatment of cerebral vascular insufficiency. Ritodrine
is a short-acting β2-ARs agonist and is also used to suppress threatened abortion.
Butaxamine/butoxamine is a selective antagonist to β2-ARs and has no clinical use. It
is used for experimentation where selective blocking of β2-ARs is required to estimate
the effect of other receptors. In contrast to cardioselective β-AR blocker, nonselective
β-blockers are widely used to treat hypertension, myocardial infraction, cardiac
arrhythmia, glaucoma, angina pectoris, CHF, atrial fibrillation etc. The blockade of
facilitatory effect of presynaptic β-ARs on noradrenaline release may also contribute to
the antihypertensive effect. Propanolol is the classical example as the nonselective
category of β-AR blocker. Prior to treating patients with β-blocker, associated adverse
effects of these drugs should be considered, viz. bronchoconstriction for patients with
airway disease like asthmatics, or obstructive pulmonary disease; bradycardia leading
to heart block for coronary disease patients; adequate cardiac output for cardiac failure
patients to impart a degree of sympathetic drive to the heart; hypoglycemia for diabetic
patients; and cold extremities due to block of beta receptor mediated vasodilation in
cutaneous vessels. Topical β-blockers are the first-choice drugs for open angle glau-
coma because these drugs lower intraocular tension by reducing aqueous formation
without affecting pupil size, tone of ciliary muscle, or outflow facility. Timolol is a
nonselective β-AR blocker. It has no local anesthetic or intrinsic sympathomimetic
activity. Cartiolol is a β blocker with intrinsic sympathomimetic activity. It can also act
as HT1A and 5-HT1B receptor antagonist. It has the ability to preserve ocular perfusion
and increase retinal blood flow. This may prevent optic nerve damage independent of
intraocular tension reduction. Hydroxycarteolol is the active metabolite of carteolol.
Levobunolol is an alternative to timolol for the management of ocular hypertension
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 131
and open-angle glaucoma. Metipranolol and mepindolol are also similar to timolol but
have different corneal anesthetic character. Befunolol is a β-AR blocker with intrinsic
sympathomimetic activity. This also acts as a β-ARs partial agonist. Pindolol is a
nonselective β-ARs blocker. It preferentially blocks 5-HT1A receptor as well as acts as
a weak partial agonist to this receptor. It also shows intrinsic sympathomimetic activity
and membrane-stabilizing effects. Tertatolol also displays similar action like
propanolol and pindolol to serotonin receptor (Langlois et al. 1993). Carvedilol is a
nonselective β-AR blocker as well as α1 blocker. Both S() and R(+) enantiomers are
responsible for later action whereas S() enantiomer is responsible for former action
(Ruffolo et al. 1990). Like carvedilol, labetalol also possesses α1 and β-ARs antago-
nistic activity. It has intrinsic sympathomimetic activity as well as membrane
stabilizing activity (Craig and Stitzel 2004; Mottram and Erickson 2009). Nebivolol
is generally categorized as a nonselective β-AR antagonist, but this unique drug shows
cardioselectivity at a dose of 5 mg and loses β1-ARs selectivity at a dose of above
10 mg (Nuttall et al. 2003). Moreover, nebivolol is devoid of cardioselectivity in
patients who are genetically poor metabolizer or who are coadministering this agent
with CYP2D6 inhibitors (Nuttall et al. 2003). Alprenolol is a nonselective β-AR
blocker with additional 5-HT1A and 5-HT1B receptor antagonist (Langlois et al.
1993). This is used for the treatment of angina pectoris (Hickie 1970). The activity
as well as application of oxprenolol is closely related to alprenolol (Langlois et al.
1993). Alprenoxime is a prodrug to alprenolol and also block β-ARs (Prokai et al.
1995). Adimolol is a nonselective α1, α2, and β-AR antagonist (Palluk et al. 1986).
Bevantolol is a β-AR blocker as well as a calcium channel blocker (Vaughan Williams
1987). Bopindolol is a β-adrenergic blocker which acts as a prodrug for its active
metabolite 4-(3-t-butylamino-2-hydroxypropoxy)-2-methylindole (Nagatomo et al.
2001). Bufuralol is a potent β-AR antagonist with partial agonist activity (Pringle
et al. 1986). Bupranolol is a nonselective β-AR blocker with strong membrane
stabilizing activity but devoid of intrinsic sympathomimetic activity. Sotalol is a
nonselective β-AR blocker with both class II and class III antiarrhythmic properties.
Like other nonselective blockers, it has also potential side effects but used generally to
treat abnormal heart rhythms associated with ventricular arrhythmias, atrial fibrillation,
or atrial flutter (Bertrix et al. 1986). Nipradilol is a β-AR antagonist and also acts as a
nitric oxide donor (Inoue et al. 2011). Medroxalol is a dual inhibitor for both α and β
ARs. Flestolol is a short-acting β-AR antagonist. Other than those mentioned above,
here are some β-AR blockers which have been developed for desired therapeutic
applications with lesser side effects like adaprolol, alfurolol, amosulalol, ancarolol,
arnolol, bornaprolol, brefonalol, bucumolol, bufetolol, bunitrolol, Butidrine,
carpindolol, cicloprolol, cinamolol, cloranolol, cyanopindolol, dalbraminol, ecastolol,
epanolol, ericolol, ersentilide, exaprolol, eugenodilol, falintolol, indenolol,
indopanolol, isoxaprolol, levomoprolol, moprolol, nadolol, nadoxolol, nifenalol,
pacrinolol, pafenolol, pamatolol, pargolol, primidolol, procinolol, ridazolol,
ronactolol, soquinolol, spirendolol, sulfinalol, talinolol, tazolol, tienoxolol, tilisolol,
tiprenolol, tolamolol, toliprolol, xibenolol, and xipranolol.
132 B. Gorain et al.
4.2.2.3 b3 Adrenoceptors
The existence of β3 ARs was first revealed in 1984, where the author reported the
existing β ARs (β1 and β2) are not specific to the novel β-adrenergic ligands (BRL
35135A, BRL 33725A, BRL 26830A). These ligands were shown to produce
remarkable anti-obesity actions on experimental animals with severe diabetes and
obesity (Arch et al. 1984). Later, this novel AR was cloned by Emorine and team
(Emorine et al. 1989).
Structural Biology
This β3 ARs, similar to β1 and β2, belong to serpentine seven transmembrane
GPCRs. Each segment of the transmembrane consists of 22–28 amino acids, with
three extracellular and three intracellular loops. Thus, altogether, these β3 receptors
contain 396 amino acids, where the C-terminal is intracellular and the N-terminal is
extracellular. This N-terminal ending is glycosylated, whereas the intracellular
C-terminal ending is phosphorylated by the action β-receptor kinases or PKA
(Coman et al. 2009). The interaction of specific ligands to this receptor and its
subsequent activity depend on the disulfide linkage between the two amino acids
(Cys110 and Cys361) in the second and third extracellular loops (Coman et al.
2009).
Additionally, four transmembrane domains are also reported crucial for the
interaction between receptors and ligands, such as transmembrane 3, 4, 5, and
6. Further, transmembrane 2 and 7 are associated with the activation of G protein
to promote the formation of a second messenger (Skeberdis 2004).
Receptor Modulators
Although β3-AR modulators along with noradrenaline-serotonin uptake inhibitor
have been focused towards the treatment of obesity, there is no β3-AR-selective
agonist being approved so far for this purpose currently (Jesudason et al. 2011).
Alternatively, the activation of β3-ARs through brown adipose tissue recently focus
on moderate sleep, eating habit and metabolic responses (Szentirmai and Kapás
2017). Several β3-ARs agonists, such as vibegron and mirabegron, are presently
approved by regulatory agencies and introduced clinically to overcome problems
associated with overactive bladder syndrome, where solabegron and ritobegron are
still under clinical investigation towards approval (Warren et al. 2016; Schena and
Caplan 2019). Amibegron has also crossed the barrier of laboratory, but
discontinued during progression in clinical research for application as antidepressant
and antianxiolytic (Schena and Caplan 2019).
134 B. Gorain et al.
4.2.3 Conclusion
References
Abel J (1899) On epinephrin, the active constituent of the suprarenal capsule and its compounds.
Am Physiol Soc 3–4:iii–iiv
Adami M, Coruzzi G, Sotirov E et al (2003) Pharmacological evidence for beta3 adrenoceptors in
the control of rat gastric acid secretion. Dig Dis Sci 48:334–339
Addison T (1855) On the constitutional and local effects of disease of the supra-renal capsules. In:
1st edn. Samuel Highley, London, pp 1793–1860
Ahlquist RP (1948) A study of the ADRENOTROPIC receptors. Am J Physiol Content
153:586–600. https://doi.org/10.1152/ajplegacy.1948.153.3.586
Altman JD, Trendelenburg AU, MacMillan L et al (1999) Abnormal regulation of the sympathetic
nervous system in α 2A -adrenergic receptor knockout mice. Mol Pharmacol 56:154–161
Amobi NIB, Guillebaud J, Kaisary AV et al (2002) Discrimination by SZL49 between contractions
evoked by noradrenaline in longitudinal and circular muscle of human vas deferens. Br J
Pharmacol 136:127–135
Arch JR, Kaumann AJ (1993) Beta 3 and atypical beta-adrenoceptors. Med Res Rev 13:663–729
Arch JRS, Ainsworth AT, Cawthorne MA et al (1984) Atypical β-adrenoceptor on brown
adipocytes as target for anti-obesity drugs. Nature 309:163–165
Awatramani GB, Price GD, Trussell LO (2005) Modulation of transmitter release by presynaptic
resting potential and background calcium levels. Neuron 48:109–121
Babin-Ebell J, Gliese M (1995) Extraneuronal uptake of noradrenaline in human tissue (uptake2).
Heart Vessel 10:151–153
Baez M, Garg D, Jallad N, Weidler D (1986) Antihypertensive effect of doxazosin in hypertensive
patients: comparison with atenolol. Br J Clin Pharmacol 21:63S–67S
Baker JG (2005) The selectivity of β -adrenoceptor antagonists at the human β 1, β 2 and β
3 adrenoceptors. Br J Pharmacol 144:317–322
Baker JG, Gardiner SM, Woolard J et al (2017) Novel selective β 1 -adrenoceptor antagonists for
concomitant cardiovascular and respiratory disease. FASEB J 31:3150–3166
Basil B, Jordan R (1982) Pharmacological properties of diacetolol (M&B 16,942), a major
metabolite of acebutolol. Eur J Pharmacol 80:47–56
Beridze N, Frishman WH (2012) Vascular Ehlers-Danlos syndrome. Cardiol Rev 20:4–7
Berkowitz DE, Nardone NA, Smiley RM et al (1995) Distribution of β3-adrenoceptor mRNA in
human tissues. Eur J Pharmacol Mol Pharmacol 289:223–228
Berlan M, Galitzky J, Bousquet-Melou A et al (1994) Beta-3 adrenoceptor-mediated increase in
cutaneous blood flow in the dog. J Pharmacol Exp Ther 268:1444–1451
Bertrix L, Timour-Chah Q, Lang J et al (1986) Protection against ventricular and atrial fibrillation
by sotalol. Cardiovasc Res 20:358–363
Björklund M, Siverina I, Heikkinen T et al (2001) Spatial working memory improvement by an α2-
adrenoceptor agonist dexmedetomidine is not mediated through α2C-adrenoceptor. Prog
Neuropsychopharmacol Biol Psychiatry 25:1539–1554
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 135
Cotecchia S, Schwinn DA, Randall RR et al (1988) Molecular cloning and expression of the cDNA
for the hamster alpha 1-adrenergic receptor. Proc Natl Acad Sci 85:7159–7163
Craig CR, Stitzel RE (2004) Modern pharmacology with clinical applications, 6th edn. Lippincott
Williams & Wilkins, Philadelphia
Daly CJ, Deighan C, McGee A et al (2002) A knockout approach indicates a minor vasoconstrictor
role for vascular α 1B -adrenoceptors in mouse. Physiol Genomics 9:85–91
Daniels DV, Gever JR, Jasper JR et al (1999) Human cloned alpha1A-adrenoceptor isoforms
display alpha1L-adrenoceptor pharmacology in functional studies. Eur J Pharmacol
370:337–343
Danner I, Escande D, Gauthier C (2001) Beta(3)-adrenoceptors control cl() conductance in rabbit
nasal epithelium. Eur J Pharmacol 422:203–207
Daubner SC, Le T, Wang S (2011) Tyrosine hydroxylase and regulation of dopamine synthesis.
Arch Biochem Biophys 508:1–12
Davenport HW (1982) Epinephrin(e). Physiologist 25:76–82
Day HE, Campeau S, Watson SJ, Akil H (1997) Distribution of alpha 1a-, alpha 1b- and alpha
1d-adrenergic receptor mRNA in the rat brain and spinal cord. J Chem Neuroanat 13:115–139
Deng CY, Lin SG, Zhang WC et al (2006) Esmolol inhibits Na(+) current in rat ventricular
myocytes. Methods Find Exp Clin Pharmacol 28:697–702
Di Cesare ML, Micheli L, Crocetti L et al (2017) α2 adrenoceptor: a target for neuropathic pain
treatment. Mini Rev Med Chem 17:95–107
Docherty JR (2010) Subtypes of functional α1-adrenoceptor. Cell Mol Life Sci 67:405–417
Dogrul A, Coskun I, Uzbay T (2006) The contribution of Alpha-1 and Alpha-2 adrenoceptors in
peripheral Imidazoline and adrenoceptor agonist-induced nociception. Anesth Analg
103:471–477
Easson LH, Stedman E (1933) Studies on the relationship between chemical constitution and
physiological action. Biochem J 27:1257–1266
Eltze M (1996) Functional evidence for an α1B-adrenoceptor mediating contraction of the mouse
spleen. Eur J Pharmacol 311:187–198
Emorine L, Marullo S, Briend-Sutren M et al (1989) Molecular characterization of the human beta
3-adrenergic receptor. Science (80-) 245:1118–1121
Fagura MS, Lydford SJ, Dougall IG (1997) Pharmacological classification of α 1 -adrenoceptors
mediating contractions of rabbit isolated ear artery: comparison with rat isolated thoracic aorta.
Br J Pharmacol 120:247–258
Frances Davies M, Tsui J, Flannery JA et al (2004) Activation of α2 adrenergic receptors suppresses
fear conditioning: expression of c-Fos and phosphorylated CREB in mouse amygdala.
Neuropsychopharmacology 29:229–239
Fulton B, Wagstaff AJ, Sorkin EM (1995) Doxazosin. An update of its clinical pharmacology and
therapeutic applications in hypertension and benign prostatic hyperplasia. Drugs 49:295–320
Gaddum JH, Holzbauer M (1957) Adrenaline and noradrenaline. Vitam Horm 15:151–203. https://
doi.org/10.1016/S0083-6729(08)60510-5
Gauthier C, Tavernier G, Charpentier F et al (1996) Functional beta3-adrenoceptor in the human
heart. J Clin Invest 98:556–562
Gingrich JA, Caron MG (1993) Recent advances in the molecular biology of dopamine receptors.
Annu Rev Neurosci 16:299–321
Goldstein DS (2010) Adrenaline and noradrenaline. In: Encyclopedia of life sciences. Wiley,
Chichester, pp 1–9
Gomi T, Ikeda T, Ikegami F (1997) Beneficial effect of alpha-blocker on hemorheology in patients
with essential hypertension. Am J Hypertens 10:886–892
Graaf PH, Shankley NP, Black JW (1996) Analysis of the effects of α1-adrenoceptor antagonists on
noradrenaline-mediated contraction of rat small mesenteric artery. Br J Pharmacol
118:1308–1316
4 Pharmacology of Adrenaline, Noradrenaline, and Their Receptors 137
Graefe K-H, Bönisch H (1988) The transport of amines across the axonal membranes of noradren-
ergic and dopaminergic Neurones. In: Trendelenburg U, Weiner N (eds) Catecholamines
I. Handbook of experimental pharmacology. Springer, Berlin, pp 193–245
Gray K, Short J, Ventura S (2008) The alpha1A-adrenoceptor gene is required for the alpha1L-
adrenoceptor-mediated response in isolated preparations of the mouse prostate. Br J Pharmacol
155:103–109
Griffith RK (2003) Adrenergics and adrenergic-blocking agents. In: Burger’s medicinal chemistry
and drug discovery. Wiley, Chichester
Haapalinna A, Leino T, Heinonen E (2003) The alpha 2-adrenoceptor antagonist atipamezole
potentiates anti-parkinsonian effects and can reduce the adverse cardiovascular effects of
dopaminergic drugs in rats. Naunyn Schmiedeberg’s Arch Pharmacol 368:342–351
Hancock AA, Kyncl JJ, Martin YC, DeBernardis JF (1988) Differentiation of alpha-adrenergic
receptors using pharmacological evaluation and molecular modeling of selective adrenergic
agents. J Recept Res 8:23–46
Hickie JB (1970) Alprenolol (&aptin&) in angina pectoris. A double-blind multicentre trial. Med J
Aust 2:268–272
Hieble JP, Bylund DB, Clarke DE et al (1995) International Union of
Pharmacology. X. Recommendation for nomenclature of alpha 1-adrenoceptors: consensus
update. Pharmacol Rev 47:267–270
Hillarp N-Å, Hokfelt B (1955) Histochemical demonstration of noradrenaline and adrenaline in the
adrenal medulla. J Histochem Cytochem 3:1–5
Hom GJ, Forrest MJ, Bach TJ et al (2001) Beta(3)-adrenoceptor agonist-induced increases in
lipolysis, metabolic rate, facial flushing, and reflex tachycardia in anesthetized rhesus monkeys.
J Pharmacol Exp Ther 297:299–307
Horinouchi T, Tanaka Y, Koike K (2002) [Beta 3-adrenoceptor-mediated relaxation of Guinea-pig
gastric funds smooth muscle: cAMP-independent characteristics and a primary role of
4-aminopyridine-sensitive voltage-dependent K+ (Kv) channels]. Nihon Yakurigaku Zasshi
120:109P–111P
Hosoda C, Koshimizu T-A, Tanoue A et al (2004) Two 1-adrenergic receptor subtypes regulating
the vasopressor response have differential roles in Blood pressure regulation. Mol Pharmacol
67:912–922
Hunter JC, Fontana DJ, Hedley LR et al (1997) Assessment of the role of alpha2-adrenoceptor
subtypes in the antinociceptive, sedative and hypothermic action of dexmedetomidine in
transgenic mice. Br J Pharmacol 122:1339–1344
Ikeshita K, Nishikawa K, Toriyama S et al (2008) Landiolol has a less potent negative inotropic
effect than esmolol in isolated rabbit hearts. J Anesth 22:361–366
Inoue K, Kei Noguchi K, Masumoto M, Wakakura M (2011) Effect of five years of treatment with
nipradilol eye drops in patients with normal tension glaucoma. Clin Ophthalmol 5:1211–1216
Ishide T (2002) Denopamine, a selective beta1-receptor agonist and a new coronary vasodilator.
Curr Med Res Opin 18:407–413
Iversen LL (1971) Role of transmitter uptake mechanisms in synaptic neurotransmission. Br J
Pharmacol 41:571–591
Jaillon P, Drici M (1989) Recent antiarrhythmic drugs. Am J Cardiol 64:65J–69J
Janumpalli S, Butler LS, MacMillan LB et al (1998) A point mutation (D79N) of the alpha2A
adrenergic receptor abolishes the antiepileptogenic action of endogenous norepinephrine. J
Neurosci 18:2004–2008
Jesudason CD, Baker JE, Bryant RD et al (2011) Combination of a Beta adrenoceptor modulator
and a norepinephrine-serotonin uptake inhibitor for the treatment of obesity. ACS Med Chem
Lett 2:583–586
Jinushi K, Kushikata T, Kudo T et al (2018) Central noradrenergic activity affects analgesic effect
of neuropeptide S. J Anesth 32:48–53
Johnston JP (1968) Some observations upon a new inhibitor of monoamine oxidase in brain tissue.
Biochem Pharmacol 17:1285–1297. https://doi.org/10.1016/0006-2952(68)90066-x
138 B. Gorain et al.
Morigny P, Houssier M, Mouisel E, Langin D (2016) Adipocyte lipolysis and insulin resistance.
Biochimie 125:259–266
Morilak DA, Barrera G, Echevarria DJ et al (2005) Role of brain norepinephrine in the behavioral
response to stress. Prog Neuropsychopharmacol Biol Psychiatry 29:1214–1224
Moriyama N, Nasu K, Takeuchi T et al (1997) Quantification and distribution of α 1 -adrenoceptor
subtype mRNAs in human vas deferens: comparison with those of epididymal and pelvic
portions. Br J Pharmacol 122:1009–1014
Morrow AL, Creese I (1986) Characterization of alpha 1-adrenergic receptor subtypes in rat brain: a
reevaluation of [3H]WB4104 and [3H]prazosin binding. Mol Pharmacol 29:321–330
Mottram AR, Erickson TB (2009) Toxicology in emergency cardiovascular care. In: Field JM,
Kudenchuk PJ, O’Connor R, Terry V (eds) The textbook of emergency cardiovascular care and
CPR. Lippincott Williams & Wilkins, Philadelphia, pp 443–452
Müller P, Schier AF (2011) Extracellular movement of signaling molecules. Dev Cell 21:145–158
Muramatsu I, Ohmura T, Kigoshi S et al (1990) Pharmacological subclassification of α1-
adrenoceptors in vascular smooth muscle. Br J Pharmacol 99:197–201
Nagatomo T, Hosohata Y, Ohnuki T et al (2001) Bopindolol: pharmacological basis and clinical
implications. Cardiovasc Drug Rev 19:9–24
Nakajima D, Negoro N, Nakaboh A et al (2006) Effectiveness of low dose denopamine, a β1-
adrenoceptor agonist, in a patient with vasospastic angina refractory to intensive medical
treatment. Int J Cardiol 108:281–283
Nakamura S, Taniguchi T, Suzuki F et al (1999) Evaluation of alpha1-adrenoceptors in the rabbit
iris: pharmacological characterization and expression of mRNA. Br J Pharmacol
127:1367–1374
Nguyen V, Tiemann D, Park E, Salehi A (2017) Alpha-2 Agonists. Anesthesiol Clin 35:233–245
Nicholas AP, Pieribone V, Hökfelt T (1993) Distributions of mRNAs for alpha-2 adrenergic
receptor subtypes in rat brain: an in situ hybridization study. J Comp Neurol 328:575–594
Nishio R, Matsumori A, Shioi T et al (1998) Denopamine, a beta1-adrenergic agonist, prolongs
survival in a murine model of congestive heart failure induced by viral myocarditis: suppression
of tumor necrosis factor-alpha production in the heart. J Am Coll Cardiol 32:808–815
Noble AJ, Chess-Williams R, Couldwell C et al (1997) The effects of tamsulosin, a high affinity
antagonist at functional α 1A - and α 1D -adrenoceptor subtypes. Br J Pharmacol 120:231–238
Nuttall SL, Routledge HC, Kendall MJ (2003) A comparison of the beta1-selectivity of three beta1-
selective beta-blockers. J Clin Pharm Ther 28:179–186
Nyrönen T, Pihlavisto M, Peltonen JM et al (2001) Molecular mechanism for agonist-promoted
alpha(2A)-adrenoceptor activation by norepinephrine and epinephrine. Mol Pharmacol
59:1343–1354
Ohmura T, Oshita M, Kigoshi S, Muramatsu I (1992) Identification of α1-adrenoceptor subtypes in
the rat vas deferens: binding and functional studies. Br J Pharmacol 107:697–704
Okajima M, Takamura M, Taniguchi T (2015) Landiolol, an ultra-short-acting β1-blocker, is useful
for managing supraventricular tachyarrhythmias in sepsis. World J Crit Care Med 4:251–257
Oliver G, Schäfer EA (1895) The physiological effects of extracts of the suprarenal capsules. J
Physiol 18:230–276. https://doi.org/10.1113/jphysiol.1895.sp000564
Palluk R, Hoefke W, Gaida W et al (1986) Interactions of MEN 935 (adimolol), a long acting beta-
and alpha-adrenolytic antihypertensive agent, with postsynaptic alpha-adrenoceptors in differ-
ent isolated blood vessels--influence of angiotensin II. Naunyn Schmiedeberg’s Arch Pharmacol
333:277–283
Parker K, Brunton L, Goodman LS et al (2008) Goodman and Gilman’s manual of pharmacology
and therapeutics. McGraw-Hill Medical, New York, NY
Perez DM, Piascik MT, Graham RM (1991) Solution-phase library screening for the identification
of rare clones: isolation of an alpha 1D-adrenergic receptor cDNA. Mol Pharmacol 40:876–883
Perez DM, Piascik MT, Malik N et al (1994) Cloning, expression, and tissue distribution of the rat
homolog of the bovine alpha 1C-adrenergic receptor provide evidence for its classification as the
alpha 1A subtype. Mol Pharmacol 46:823–831
140 B. Gorain et al.
Strosberg AD (1993) Structure, function, and regulation of adrenergic receptors. Protein Sci
2:1198–1209. https://doi.org/10.1002/pro.5560020802
Summers RJ, Papaioannou M, Harris S, Evans BA (1995) Expression of beta 3-adrenoceptor
mRNA in rat brain. Br J Pharmacol 116:2547–2548
Suryanarayana S, von Zastrow M, Kobilka BK (1992) Identification of intramolecular interactions
in adrenergic receptors. J Biol Chem 267:21991–21994
Szentirmai É, Kapás L (2017) The role of the brown adipose tissue in β3-adrenergic receptor
activation-induced sleep, metabolic and feeding responses. Sci Rep 7:958
Szymonowicz L (1896) Die Function der Nebenniere. Pflüger, Arch 64:97–164. https://doi.org/10.
1007/BF01661663
Tagaya E, Tamaoki J, Takemura H et al (1999) Atypical adrenoceptor-mediated relaxation of
canine pulmonary artery through a cyclic adenosine monophosphate-dependent pathway. Lung
177:321–332
Takamine J (1902) The blood-pressure raising principle of the suprarenal gland. JAMA
XXXVIII:153–155. https://doi.org/10.1001/jama.1902.62480030011001c
Tamaoki J, Yamauchi F, Chiyotani A et al (1993) Atypical beta-adrenoceptor- (beta
3-adrenoceptor) mediated relaxation of canine isolated bronchial smooth muscle. J Appl Physiol
74:297–302
Teng C-M, Guh J-H, Ko F-N (1994) Functional identification of α1-adrenoceptor subtypes in
human prostate: comparison with those in rat vas deferens and spleen. Eur J Pharmacol
265:61–66
Tran TM, Friedman J, Qunaibi E et al (2004) Characterization of agonist stimulation of cAMP-
dependent protein kinase and G protein-coupled receptor kinase phosphorylation of the
2-adrenergic receptor using phosphoserine-specific antibodies. Mol Pharmacol 65:196–206
Trendelenburg U (1991) The TiPS lecture: functional aspects of the neuronal uptake of noradrena-
line. Trends Pharmacol Sci 12:334–337
Vargas HM, Gorman AJ (1995) Vascular alpha-1 adrenergic receptor subtypes in the regulation of
arterial pressure. Life Sci 57:2291–2308
Vaughan Williams EM (1987) Bevantolol: a beta-1 adrenoceptor antagonist with unique additional
actions. J Clin Pharmacol 27:450–460
Villalobos-Molina R, López-Guerrero JJ, Ibarra M (1997) Alpha 1D- and alpha 1A-adrenoceptors
mediate contraction in rat renal artery. Eur J Pharmacol 322:225–227
Vinay HK, Paul A, Goswami SS, Santani D (2002) Effect of SR 58611A, a beta-3 receptor agonist,
against experimental gastro-duodenal ulcers. Indian J Physiol Pharmacol 46:36–44
Wada Y, Aiba T, Tsujita Y et al (2016) Practical applicability of landiolol, an ultra-short-acting β1-
selective blocker, for rapid atrial and ventricular tachyarrhythmias with left ventricular dysfunc-
tion. J Arrhythm 32:82–88
Wallukat G (2002) The β-adrenergic receptors. Herz 27:683–690
Warne T, Serrano-Vega MJ, Baker JG et al (2008) Structure of a beta1-adrenergic G-protein-
coupled receptor. Nature 454:486–491
Warren K, Burden H, Abrams P (2016) Mirabegron in overactive bladder patients: efficacy review
and update on drug safety. Ther Adv drug Saf 7:204–216
Wassall RD, Teramoto N, Cunnane TC (2009) Noradrenaline. In: Encyclopedia of Neuroscience.
Academic Press, Cambridge, pp 1221–1230
Webb DJ, Fulton JD, Leckie BJ et al (1987) The effect of chronic prazosin therapy on the response
of the renin-angiotensin system in patients with essential hypertension. J Hum Hypertens
1:195–200
Wei L, Zhu Y-M, Zhang Y-X et al (2016) The α1 adrenoceptors in ventrolateral orbital cortex
contribute to the expression of morphine-induced behavioral sensitization in rats. Neurosci Lett
610:30–35
142 B. Gorain et al.
Westlund KN, Denney RM, Rose RM, Abell CW (1988) Localization of distinct monoamine
oxidase a and monoamine oxidase b cell populations in human brainstem. Neuroscience
25:439–456
White WB, Moon T (2005) Treatment of benign prostatic hyperplasia in hypertensive men. J Clin
Hypertens 7:212–217
Woodcock EA (2007) Roles of α1A- and α1B-adrenoceptors in heart: insights from studies of
genetically modified mice. In: Clinical and experimental pharmacology and physiology. Wiley,
Chichester
Wurtman RJ, Axelrod J (1966) Control of enzymatic synthesis of adrenaline in the adrenal medulla
by adrenal cortical steroids. J Biol Chem 241:2301–2305
Xie F, Petitti D, Chen W (2005) Prescribing patterns for antihypertensive drugs after the antihyper-
tensive and lipid-lowering treatment to prevent heart attack trial: report of experience in a health
maintenance organization. Am J Hypertens 18:464–469
Yu GS, Han C (1994) Role of alpha 1A- and alpha 1B-adrenoceptors in phenylephrine-induced
positive inotropic response in isolated rat left atrium. J Cardiovasc Pharmacol 24:745–752
Zhao T-J, Sakata I, Li RL et al (2010) Ghrelin secretion stimulated by 1-adrenergic receptors in
cultured ghrelinoma cells and in fasted mice. Proc Natl Acad Sci 107:15868–15873
Zuscik MJ, Chalothorn D, Hellard D et al (2001) Hypotension, autonomic failure, and cardiac
hypertrophy in transgenic mice overexpressing the α 1B -adrenergic receptor. J Biol Chem
276:13738–13743
Pharmacology of Dopamine and Its
Receptors 5
Sunpreet Kaur, Shamsher Singh, Gagandeep Jaiswal,
Sandeep Kumar, Wafa Hourani, Bapi Gorain, and Puneet Kumar
Abstract
Keywords
Abbreviations
5-HT 5-Hydroxytryptamine
AADC Aromatic amino acid decarboxylase
AC Adenyl cyclase
ACC Anterior cingulated
AD Alzheimer’s disease
ADH Alcohol dehydrogenase
ALDH Aldehyde dehydrogenase
ALLO Allopregnanolone
AMP Adenosine monophosphate
AMPA α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
BLA Basolateral amygdala
cAMP Cyclic adenosine monophosphate
COMT Catechol-O-methyl-transferase
CREB cAMP response element binding protein
D1R D1 receptor
D5R D5 receptor
DA Dopamine
DARPP-32 Dopamine- and cAMP-regulated neuronal phosphoprotein
DAT Dopamine transporters
DHX Dihydrexidine
DOPA Dihydroxyphenylalanine
DOPAC 3,4-Dihydroxyphenylacetic acid
DOPAL 3,4-Dihydroxyphenylacetaldehyde
5 Pharmacology of Dopamine and Its Receptors 145
DOPET 3,4-Dihydroxyphenylethanol
DSM-IV Diagnostic and Statistical Manual of Mental Disorders
EPS Extrapyramidal symptoms
FAD Flavin adenine dinucleotide
GABA Gamma-aminobutyric acid
GAD Generalized anxiety disorder
GPCRs G-protein-coupled receptors
GPe External globus pallidus
GPi Globus pallidus internal
Gαi Gi alpha subunit
Gαs Gs alpha subunit
HD Huntington’s disease
HVA Homovanillic acid
L-DOPS L-Dihydroxyphenylserine
MAO Monoamino oxidase
MAPK Mitogen-activated protein kinase
MS Multiple sclerosis
MSN Medium spiny neuron
NAc Nucleus accumbens
NMDA N-methyl-D-aspartate receptor
O2 Molecular oxygen
OB Olfactory bulb
OCC Orbitofrontal cortical
OCD Obsessive compulsive disorder
PD Parkinson’s disease
PFC Prefrontal cortex
PKA Protein kinase A
PKC Protein kinase C
PP1 Protein phosphatase
PRMS Progressive relapsing multiple sclerosis
PTSD Post-traumatic stress disorder
RNA Ribose nucleic acid
RRMS Relapsing remitting multiple sclerosis
SCZ Schizophrenia
SNpc Substantia nigra pars compacta
SNpr Substantia nigra pars reticulata
SPD Sensory processing disorder
SPMS Secondary progressive multiple sclerosis
STN Subthalamic nucleus
TBI Traumatic brain injury
TBZ Tetrabenazine
TH Tyrosine hydroxylase
VMAT2 Vesicular monoamine transporter 2
VTA Ventral tegmental area
146 S. Kaur et al.
5.1 Introduction
5.2 History
The classical pathway of DA biosynthesis was initially stated by Blaschko in the late
1930s. It involves two events, proceed with hydroxylation in L-tyrosine to
dihydroxyphenylalanine (DOPA) catalysed by tyrosine hydroxylase (TH) in
the presence of tetrahydrobiopterin, O2, and iron (Fe2+) as cofactors (Juárez Olguín
et al. 2016). Further, DOPA gets decarboxylated to final product DA via enzyme
aromatic amino acid decarboxylase (AADC) where pyridoxal phosphate (PP) is
present as a cofactor. The tyrosine utilized in this pathway formed by the enzyme
phenylalanine hydroxylase in the presence of molecular oxygen (O2) and
tetrahydrobiopterin as cofactors (Meiser et al. 2013) (Fig. 5.2). Two other alternative
pathways of dopamine synthesis can be described in the form of flow charts as
shown in Figs. 5.3 and 5.4.
After formation, dopamine gets readily sequestered inside the synaptic vesicles
through vesicular monoamine transporter 2 (VMAT-2) and remains stored under
slightly acidic pH to prevent oxidation of dopamine. This vesicular transport irre-
versibly inhibited by reserpine and amphetamine like drugs (Meiser et al. 2013).
Further upon excitation of dopaminergic neurons, synaptic vesicles get
degranulated, hence releasing dopamine in the synapse where it interacts with
dopamine receptors for subsequent actions and dopamine transporters (DAT)
remove excess of dopamine from synapse.
L-DOPA
Dopamine
Tyramine
Hydroxylation by
CYP2D
Dopamine (very
low quantity)
The physiological actions of dopamine are mediated by its interaction with dopa-
mine receptors that are localized over dopaminergic synapses. There are five types of
dopamine receptors categorized into two different subclasses including D1-like and
D2-like dopamine receptors (Beaulieu et al. 2015). The D1-like class includes D1
and D5 receptors (D1R and D5R), whereas D2-like class constitutes of D2, D3, and
D4 receptors (D2R, D3R, and D4R). Both classes of receptors differ in their
mechanism of action. The D1-like receptors act by coupling to Gαs that causes
adenylyl cyclase (AC) activation for cAMP production while D2-like couples to
Gαi to negatively regulate AC. Here, the D1-like receptors are found to be expressed
only postsynaptically but D2-like receptors are expressed both presynaptically and
postsynaptically.
The D1 receptors are distributed over nigrostriatal, mesolimbic, and mesocortical
areas, like the caudate-putamen (striatum), substantia nigra, olfactory bulb, amyg-
dala and at small level in the hippocampus, cerebellum, thalamic areas, and hypo-
thalamic areas (Rangel-Barajas et al. 2015). On the other side, D5 receptors are
found to be expressed at a very low level in the prefrontal cortex, premotor cortex,
cingulated cortex, entorhinal cortex, substantia nigra, hypothalamus, hippocampus,
and dentate gyrus. A small proportion is also reported over medium spiny neurons of
the caudate nucleus and nucleus accumbens.
152 S. Kaur et al.
The D2 receptors are expressed on the striatum, the substantia nigra, ventral
tegmental area, hypothalamus, cortical areas, septum, amygdala, hippocampus, the
nucleus accumbens, and the olfactory tubercle like regions (Rangel-Barajas et al.
2015). The distribution of D3 receptor is considered smaller in the limbic areas,
striatum, the substantia nigra, pars compacta, the ventral tegmental area, hippocam-
pus, septal area, and various cortical areas. Low expression of D4 receptor is
reported upon the frontal cortex, amygdala, hippocampus, hypothalamus, globus
pallidus and substantia nigra pars reticulata (Rangel-Barajas et al. 2015). Apart from
this, D1, D2, and D4 receptors have been reported in retina whereas D2 receptors
are detected in pituitary gland.
It includes subfamilies of D2, D3, and D4, whereas D2R subtypes possess
2 isoforms: the D2-short and D2-long type receptors. Most brain regions like
striatum, external globus pallidus (GPe), amygdala, cerebral cortex, hippocampus,
and pituitary possess D2 receptor and its subtypes (Mishra et al. 2018). Usually the
prefrontal, temporal, and entorhinal cortex, and the septal region along with the VTA
and SNpc of DAergic neurons show the expression of messenger RNA D2R
(Villanueva 2015). The activity of AC and the production of cAMP levels and
5 Pharmacology of Dopamine and Its Receptors 153
Fig. 5.6 Regulation of the signalling networks by DA in D1-like receptors, D2-like receptors, and
D1-D2 receptor heteromers. DA D1-like family and D2-like family receptor homodimers signal
through Gαs/olf and Gαi/o protein to regulate cyclic AMP through adenylyl cyclase (AC) activity.
D1-like receptors activate AC through Gαs/olf, thereby increasing intracellular cAMP and
stimulating PKA. D2-like receptors inhibit AC through Gαi/o, thereby suppressing cAMP and
inhibiting PKA. The DA D1-D2 receptor heterodimer signals through Gαq, phospholipase C,
enhancing the production of IP3, mobilization of intracellular calcium and of DAG with subsequent
activation of PKC
PKA are inhibited by this class of receptors (Zhang et al. 2006). Behavioural and
extrapyramidal activities are mediated by the D2-type postsynaptic receptor. D2
receptors are known as autoreceptors acting via somato dendritic auto-receptors,
which are known to decrease neuronal excitability (Chiodo and Kapatos 1992;
Lacey et al. 1987) or inhibit dopamine release by terminal auto-receptors, which
mostly reduce DA synthesis and packaging (Onali et al. 1988; Pothos et al. 1998).
DA neuronal development is due to D2 auto-receptor during the embryonic stage
(Baik 2013) and mediate various responses like cell proliferation–related pathways,
such as the mitogen-activated protein kinase (MAPK) (Yoon and Baik 2013) and
Akt (thymoma viral proto-oncogene also knows as protein kinase B) signalling
pathways (Collo et al. 2013) (Table 5.2).
The dopamine receptors are widespread across the brain and their crucial localization
makes them play a variety of actions. The presynaptic localization of D2 auto-
receptors inhibits dopamine release that decreases locomotor activity. On the other
154
D2 receptor for motivation. Alterations that occur in phasic and tonic responses
result from interaction with cortico-striatal glutaminergic synapses that modulate the
functional signalling of D1 and D2 receptors expressing GABAergic medium spiny
neurons (MSN). The striatal excitatory D1 receptors expressing MSN act through
direct pathway while D2-MSN receptors act in an inhibitory manner through indirect
pathway (Paladini and Roeper 2014). Both the ventral striatal pathways are
evidenced to participate in reward and punishment respectively.
Let cite an example of abused drug which firstly activates the D1 and D2
receptors via enhancing the dopamine release. When dopamine decreases rate
dependently, only D2 receptors remain active and justify the reward phenomenon
in patients (Zweifel et al. 2009). Moreover, the identical pathways also justify
associative learning which brings out the involvement of glutaminergic projections
emerging from the hippocampus, amygdala, and prefrontal cortex. These projections
uniquely regulate activation of D1 receptor that remains responsible for emotion-
associated learning, hippocampal-dependent learning, and cortex associative
learning. Besides this, the role of D2 receptor is confirmed in addiction as the
expression of the receptor in striatum gets downregulated on repeated exposure of
abuse drugs (Volkow and Morales 2015). The downregulation of D2 receptor in
striatum mediates the suppression of the indirect pathway which further distorts the
thalamo-cortical activity in prefrontal cortex. Moreover, decreasing availability of
D2 receptors in both dorsal and ventral regions had been reported in PFC of abused
patients specifically in anterior cingulated (ACC) and orbitofrontal cortical (OCC)
regions. Altered functioning of dopamine receptors in these two regions is observed
to be responsible for impulsive and compulsive behaviour in drug abusers. Hence, in
5 Pharmacology of Dopamine and Its Receptors 157
this way, it is the ratio and imbalance in between the D1- to D2-mediated direct and
indirect pathway that uniquely contribute to addiction, learning, and other
behavioural changes.
Specific agonists and antagonists of dopamine are available for D1 and D2 class of
dopamine. Selective agonists for D1 class include A77636, SKF38393, SKF81297,
and dihydrexidine, while D2 selective compounds include quinpirole and N-0435.
On the other side, antagonists of D1 class involve SCH23390, SCH39166, and
SKF35566, while D2 antagonism is caused by domperidone, nemonapride,
raclopride, and sulpiride (Smee and Overstreet 1976). Moreover, some compounds
show receptor-specific actions and participate in dopamine mediated functioning
as enlisted in Table 5.2.
D1 receptor family agonists and antagonists and their functions:
1. Agonists:
(a) Dihydrexidine: It is a selective D1 agonist. Dihydrexidine gives its action
through stimulating inspiratory motor output and by depressing medullary
expiratory neurons. Dihydrexidine has been used in clinical trials for study-
ing the treatment of SPD, cocaine-related disorders, and schizotypal person-
ality disorder (Lalley 2009).
(b) Fenoldopam: Fenoldopam is a fast-acting vasodilator. It is an agonist for
D1-like dopamine receptors and has a moderate affinity towards binding to
the α2-adrenoceptors. Fenoldopam is a racemic mixture and the R-isomer is
responsible for the biological activity. The R-isomer has approximately
250-fold higher affinity for D1-like receptors than does the S-isomer. It
helps lowering down the blood pressure through arteriolar vasodilation.
Thus, this agonist is also used as an antihypertensive agent (Felder et al.
1993).
(c) SKF38393: It is a selective D1 agonist. It improves motor function in PD
(Robertson et al. 1990).
(d) SKF81297: It is a selective D1/D5 receptor agonist. It is believed to reduce
long-term potentiation to prevent synaptic failure (Reavill et al. 1993).
2. Antagonists:
(a) SCH23390: It is a D1 antagonist and believed to improve motor function in
PD and has anxiolytic effects on the hippocampus, VTA (ventral tegmental
area) (Lidow et al. 1991).
(b) SCH39366: It is a classical benzazepine D1/D5 antagonist. It has been used
in human clinical trials for studying different diseases including schizophre-
nia, cocaine addiction, and obesity (Wu et al. 2005).
158 S. Kaur et al.
over cortical activity by subthalamic nuclei and globus pallidus external. It has been
reported that this system gets dysregulated in HD and loss over movement control
occurs in the form of chorea. The early symptoms of HD associate with the loss of
D2 receptors which initiate uncontrolled dance-like movements by creating a deficit
in inhibitory regulation of the dopamine system, whereas in later stages of HD, on
the other hand, the lack of D1 receptors also results in thalamus inhibition inducing
dystonia, rigidity, and akinesia-like symptoms. Furthermore, evidence-based justifi-
cation lies behind the degeneration of this system. It has been analysed that dopa-
mine treatment differently modulates NMDA and AMPA receptors. These responses
occur due to the localization of dopamine receptor on glutaminergic terminals. The
involvement of D2 receptors in glutaminergic terminals through excitotoxicity may
exacerbate neurotoxicity in HD, whereas D1 receptor itself mediates potentiation of
NMDA receptor that contributes to neurotoxicity (Chen et al. 2013). Further,
agonists of D1 enhance neurotoxicity via NMDA receptor activation whereas
the activation of D2 receptor reduces NMDA activation for neuroprotection.
Hence, D1 receptors remain neurotoxic whereas D2 receptors prove to be
neuroprotective in HD.
Moreover, cell death of dopamine neurons also proceeds by free radical produc-
tion as they remain more prone to oxidation due to their distinct features. Changes in
dopamine levels also contribute to cognitive loss in earlier HD as cholinergic
neurons reside in striatum also degenerate, thus deteriorating cognitive functions
like attention, execution, learning, and cognition (Wang et al. 2006). The initial
phase of HD reveals to be in a hyperdopaminergic state, so reducing dopamine
remains a potential treatment strategy. The only approved drug in HD is
tetrabenazine (TBZ) that inhibits vesicular monoamine transporter to reduce dopa-
mine in presynaptic vesicles which reduce chorea and other symptoms of
HD. Aripiprazole is a partial D2 receptor agonist that improves symptoms of chorea
5 Pharmacology of Dopamine and Its Receptors 163
Table 5.5 Enlisting the compounds targeting dopamine for therapeutic efficacy in HD
Drug Mechanism of action Status in HD
Tetrabenazine Vesicular monoamine transporter to reduce Clinically approved drug
dopamine level
Haloperidol Typical D2 antagonists Improves symptoms of
disease
Olanzapine, Atypical D2 antagonists Improve chorea and
risperidone behavioural disturbances
Bromocriptine D2 agonist Improves chorea and other
motor symptoms
Lisuride D2 agonist Provides symptomatic relief
Aripiprazole Partial agonist of D2 Improves chorea but
cognition not improved
L-DOPA Dopamine agonist Provides relief over rigidity
Pridopidine DA stabilizer Improves motor dysfunction
SCH23390 D1 antagonist Shows positive result in
animal studies
SKF38393 D1 agonist Shows positive result in
animal studies
Haloperidol D2 antagonist Shows positive result in
animal studies
Quinpirole D2 agonist Shows positive result in
animal studies
Table 5.6 Enlisting the compounds targeting dopamine for therapeutic efficacy in AD
Drug Mechanism of action Status in AD
Levodopa DA precursor Improves memory and decreases synaptic
plasticity in hippocampus
Selegiline MAO-B inhibitor Improves memory and decreases synaptic
plasticity in hippocampus
Allopregnanolone Positive modulator of Increases survival of dopaminergic
(ALLO) GABAB neurons and improves cognitive deficits
Vindeburnol Promotes catecholamine Improves memory and concentration in
synthesis AD studies
L-DOPS Promotes catecholamine Improves memory and concentration in
synthesis AD studies
S33138, FP17141 D3 receptor antagonist Reverse cognitive decline
SKF81297 Selective D1/D5 receptor Reduces long-term potentiation to prevent
agonist synaptic failure
PD168077 D4 receptor agonist Enhances working memory and attention
Dimebon Increases dopamine level in Effective in mild to moderate AD
the brain
Rimonabant CB1 receptor antagonist Improves social and working memory
Increases dopamine level
Methylphenidate Dopamine reuptake Improves neuropsychiatric symptoms
inhibitor
ITI-007 Dopamine receptor Improves neuropsychiatric symptoms
modulator
Rasagiline Monoamine oxidase B Enhances cognition
inhibitor
Rotigotine Dopamine agonist Improves executive function in AD
proceed along with cognitive impairments like working memory and attention
deficits (Robertson et al. 1990). The pathogenesis of SCZ strikes the neurochemical
alterations occurring in patients with SCZ. It includes dopamine theory, glutamate
theory, and other neurochemicals alterations like serotonin dysfunction (Laruelle
2014). As dopamine theory always remains a major contributor in pathogenesis, its
role in SCZ does not remain controversial.
Initially proposed dopamine hypothesis postulates that hyperactivity of dopamine
neurotransmission results in schizophrenic symptoms. This theory was put forward
on the basis of evidence that the administration of dopamine releaser amphetamine
and dopamine enhancer levodopa potentially exacerbate symptoms of schizophrenia
(Brisch et al. 2014). On the other side, treatment drugs against enhanced dopa-
mine levels decrease the activity of dopamine and improve therapeutic reliability on
antipsychotic drugs including D2 antagonists. The mesostriatal dopamine system
remains a key target of these drugs confirmed by blockage of high concentrations of
D2 receptors that also contribute to extrapyramidal side effects. Later revisions to
classical dopamine theory provided a new finding (da Silva Alves et al. 2008). This
new evidence suggests that amphetamine does not exhibit all symptoms of schizo-
phrenia but only exacerbates positive symptoms along with the improvement in
5 Pharmacology of Dopamine and Its Receptors 167
Table 5.7 Enlisting the compounds targeting dopamine for therapeutic efficacy in
schizophrenia (SCZ)
Typical antipsychotics Atypical antipsychotics
(first-generation (second-generation Drugs under preclinical and
antipsychotics) antipsychotics) clinical trials
Chlorpromazine Aripiprazole ITI-007 (presynaptic partial D2
agonist and postsynaptic D2
antagonist)
Fluphenazine Clozapine RP5063 (partial D2, D3, and D4
agonist)
Haloperidol Lurasidone SKF-38393, SKF-83959, SPD-451
(partial D1 agonist)
Perphenazine Olanzapine DAR-0100A
Thioridazine Paliperidone SKF-81297
Thiothixene Quetiapine A-77636
Risperidone ABT-431
168 S. Kaur et al.
Table 5.8 Enlisting the compounds targeting dopamine for therapeutic efficacy in anxiety
Mechanism of
Drugs action Effects observed (region specific)
Apomorphine D1/D2 receptor Anxiolytic effects (amygdala), anxiogenic effects
agonist (hippocampus)
SCH23390 D1 antagonist Anxiolytic effects (BLA, VTA, NAc), anxiogenic effects
(hippocampus, amygdala)
Quinpirole D2 agonist Anxiolytic effects (VTA), anxiogenic effects
(hippocampus, BLA)
Sulpiride D2 antagonist Anxiolytic effects (BLA), anxiogenic effects (amygdala)
Raclopride D2 receptor Anxiolytic effects (BLA), anxiogenic effects (CeA)
antagonist
Eticlopride D2 receptor Anxiolytic effects (CeA)
antagonist
SKF38393 D1 agonist Anxiogenic effects (BLA, hippocampus)
2015). Thus, VTA remains a main unit in regulating anxiety and it is regulated by
excitatory and inhibitory inputs.
Moreover, the hippocampus not only modulates learning and memory but also
involved in fear and anxiety-like behavioural disorders. There are two different
regions of the hippocampus including ventral hippocampus and dorsal hippocampus
that differentially precede their functions. The ventral hippocampus involves fear
and anxiety due to its projections to the prefrontal cortex whereas the dorsal just
regulates memory and learning-like functions (Bannerman et al. 2004). This contri-
bution is further confirmed by lesions of the ventral hippocampus that contributes to
anxiety-like symptoms. The mesolimbic dopaminergic projections from ventral
tagmental area and SNc when gets affected from plasticity also contribute to anxiety.
On this basis several agonists and antagonists are going to target the treatment of
anxiety; some of them are enlisted in Table 5.8.
Epilepsy is the most widespread neurological disorder that initiates with uncontrol-
lable excitatory neurotransmission. It affects 1–2% of the world’s population and
features recurrent epileptic seizures. Seizures may affect localized areas (“focal” or
“partial” seizures) or spread throughout the whole cerebral hemisphere
(“generalized” seizures). The predisposing factor behind epilepsy is the hyperactiv-
ity persisting in different brain regions that involve imbalance between excitatory
and inhibitory impulses (Stafstrom and Carmant 2015). Currently most of
the approved drugs in epilepsy target this imbalance by upregulating the GABAergic
system in the brain. Whether, GABA/Glutamate remains a main target for the
development of drug therapy of epilepsy but recently growing evidence strengthens
the role of other neurotransmitters and neuropeptide in the epileptic brain.
170 S. Kaur et al.
Table 5.10 Enlisting the compounds targeting dopamine for therapeutic efficacy in TBI
Drugs Mechanism of action Pharmacological effects in TBI
Methylphenidate Increases dopamine synthesis Improves cognition, working memory,
and attention
Bromocriptine D2 receptor agonist Improves cognition
Atomoxetine Increases dopamine Improves cognition and attention
Guanfacine Increases dopamine Improves cognition and attention
Levodopa Dopamine agonist Improves cognition and attention
Methamphetamine Increases dopamine Improves memory and cognition
Amantadine Increases dopamine Improves depression symptoms
Selegiline Monoamine-oxidase-B Prevents excitotoxicity
(MAO-B) inhibitor
Rasagiline Monoamine-oxidase-B Prevents excitotoxicity
(MAO-B) inhibitor
Pramipexole Dopamine agonist Prevents excitotoxicity
Ropinirole Dopamine agonist Prevents excitotoxicity
Buproprion Increases dopamine Reduces neuroinflammation
Table 5.11 Enlisting the compounds targeting dopamine for therapeutic efficacy in MS
Drugs Mechanism of action Pharmacological effects in MS
Bromocriptine Dopamine agonist Improves chronic fatigue and other symptoms
Methylphenidate Dopamine agonist Improves chronic fatigue symptoms
Amantadine Dopamine agonist Improves chronic fatigue symptoms
Risperidone D2 receptor like Decrease spinal cord lesions and autoimmunity
antagonist
Phenelzine MAO-B inhibitor Improves MS symptoms
Fenoldopam D1-like receptor agonist Decreases autoimmunity and improves MS
symptoms
Dopamine Dopamine agonist Decreases autoimmunity and improves MS
symptoms
L-DOPA Dopamine precursor Decreases autoimmunity
Pergolide Dopamine agonist Decreases autoimmunity
Haloperidol D2 antagonist Decreases autoimmunity
Pimozide D2 antagonist Decreases autoimmunity
Fluoxetine D2 blocker Decreases autoimmunity and neuroinflammation
Domperidone D2 antagonist Decreases autoimmunity and neuroinflammation
The recent advancements in dopamine are fully contributed by eventual set of high
pace discoveries occurred in the research field. The presence of dopamine in the brain
was first reported in the 1960s but gets revolutionized when the mystery of parkin-
sonism got unlocked. Some Swedish works found it to be responsible for extrapyra-
midal symptoms of PD. Afterward scientific discoveries were mostly devoted to
possible roles of catecholamine synthesis and dopamine until levodopa was
introduced. The following years were remarkably well known for dopaminergic
pathways and their contributory role in various disorders. In the 1980s,
advancements in techniques and modern concepts of discovery highly evolved the
5 Pharmacology of Dopamine and Its Receptors 175
5.17 Conclusion
Dopamine regulates many functions through its receptor signalling which are com-
plex and may also depend on cellular protein like kinases or other enzymes. The role
of dopamine has been well studied extensively in Parkinson’s, Alzheimer’s, and
Huntington’s disease but its role is also widely expressed in various other neurologi-
cal disorders like epilepsy, multiple sclerosis, schizophrenia, anxiety, and traumatic
brain injury. In this chapter, various roles of dopamine and dopaminergic receptors
have been brightly highlighted. Several disorders demand a lot of research on
the dopaminergic system for various therapeutic approaches. Hence, the high pace
176 S. Kaur et al.
References
Arias-Carrión O, Stamelou M, Murillo-Rodríguez E, Menéndez-González M, Pöppel E (2010)
Dopaminergic reward system: a short integrative review. Int Arch Med 3(1):24
Arnaud-Batista FJ, Peruchetti DB, Abreu TP, do Nascimento NR, Malnic G, Fonteles MC, Caruso-
Neves C (2016) Uroguanylin modulates (Na++ K+) ATPase in a proximal tubule cell line:
interactions among the cGMP/protein kinase G, cAMP/protein kinase a, and mTOR pathways.
Biochim Biophys Acta 1860(7):1431–1438
Atcherley CW et al (2015) The coaction of tonic and phasic dopamine dynamics. Chem Commun
51(12):2235–2238. Royal Society of Chemistry
Baik JH (2013) Dopamine signaling in food addiction: role of dopamine D2 receptors. BMB Rep 46
(11):519
Bannerman DM et al (2004) Regional dissociations within the hippocampus—memory and anxiety.
Neurosci Biobehav Rev 28(3):273–283. Elsevier
Barger G, Dale HH (1910) Chemical structure and sympathomimetic action of amines. J Physiol 41
(1–2):19–59. Wiley Online Library
Beaulieu J, Espinoza S, Gainetdinov RR (2015) Dopamine receptors–IUPHAR review 13. Br J
pharmacol 172(1):1–23. Wiley Online Library
Beierholm U et al (2013) Dopamine modulates reward-related vigor. Neuropsychopharmacology.
38(8):1495. Nature Publishing Group
Bloom FE, Costa E, Salmoiraghi GC (1965) Anesthesia and the responsiveness of individual
neurons of the caudate nucleus of the cat to acetylcholine, norepinephrine and dopamine
administered by microelectrophoresis. J Pharmacol Exp Ther 150(2):244–252. ASPET
Bozzi Y, Borrelli E (2013) The role of dopamine signaling in epileptogenesis. Front Cell Neurosci
7:157. Frontiers
Braak H, Del Tredici K (2015) The preclinical phase of the pathological process underlying
sporadic Alzheimer’s disease. Brain 138(10):2814–2833. Oxford University Press
Bressan RA et al (2003) Optimizing limbic selective D2/D3 receptor occupancy by risperidone: a
[123I]-epidepride SPET study. J Clin Psychopharmacol 23(1):5–14. LWW
Brisch R et al (2014) The role of dopamine in schizophrenia from a neurobiological and evolution-
ary perspective: old fashioned, but still in vogue. Front Psychiatry 5:47. Frontiers
Brown JH, Makman MH (1972) Stimulation by dopamine of adenylate cyclase in retinal
homogenates and of adenosine-30 : 50 -cyclic monophosphate formation in intact retina. Proc
Natl Acad Sci U S A 69(3):539–543. National Acad Sciences
Bunzow JR et al (1988) ‘Cloning and expression of a rat D 2 dopamine receptor cDNA’, Nature
336(6201):783–787
Carlsson A (1959) The occurrence, distribution and physiological role of catecholamines in the
nervous system. Pharmacol Rev 11(2):490–493. ASPET
Carlsson A, Falck B, Hillarp N-Å (1962) Cellular localization of brain monoamines. Acta Physiol
Scand Suppl 56(196):1
Chaudhuri A, Behan PO (2000) Fatigue and basal ganglia. J Neurol Sci 179(1–2):34–42. Elsevier
Chen JY et al (2013) Dopamine imbalance in Huntington’s disease: a mechanism for the lack of
behavioral flexibility. Front Neurosci 7:114. Frontiers
Chen Y-H et al (2017) Impact of traumatic brain injury on dopaminergic transmission. Cell Transpl
26(7):1156–1168. SAGE Publications Sage CA: Los Angeles, CA
Chiodo LA, Kapatos G (1992) Membrane properties of identified mesencephalic dopamine neurons
in primary dissociated cell culture. Synapse 11(4):294–309. Wiley Online Library
178 S. Kaur et al.
Haber SN (2016) Corticostriatal circuitry. In: Neuroscience in the 21st century. Springer,
New York, NY, pp 1–21
Hasbi A, O’Dowd BF, George SR (2011) Dopamine D1-D2 receptor heteromer signaling pathway
in the brain: emerging physiological relevance. Mol Brain 4(1):26. BioMed Central
Hirvonen J et al (2006) Striatal dopamine D1 and D2 receptor balance in twins at increased genetic
risk for schizophrenia. Psychiatry Res 146(1):13–20. Elsevier
Hornykiewicz O (1958) The action of dopamine on the arterial blood pressure of the guinea-pig. Br
J Pharmacol Chemother 13(1):91–94. Wiley Online Library
Hornykiewicz O (2002) L-DOPA: from a biologically inactive amino acid to a successful therapeu-
tic agent. Amino Acids 23(1–3):65–70. Springer
Hornykiewicz O (2006) The discovery of dopamine deficiency in the Parkinsonian brain. In:
Parkinson’s disease and related disorders. Springer, New York, NY, pp 9–15
Howes O, McCutcheon R, Stone J (2015) Glutamate and dopamine in schizophrenia: an update for
the 21st century. J Psychopharmacol 29(2):97–115. Sage Publications Sage UK: London,
England
Irwin DJ, Trojanowski JQ (2013) Many roads to Parkinson’s disease neurodegeneration: head
trauma-a road more traveled than we know? Mov Disord 28(9):1167. NIH Public Access
Jahn H (2013) Memory loss in Alzheimer’s disease. Dialogues Clin Neurosci 15(4):445. Les
Laboratoires Servier
Jenkins PO, Mehta MA, Sharp DJ (2016) Catecholamines and cognition after traumatic brain
injury. Brain 139(9):2345–2371. Oxford University Press
Juárez Olguín H et al (2016) The role of dopamine and its dysfunction as a consequence of
oxidative stress. Oxid Med Cell Longev 2016. https://doi.org/10.1155/2016/9730467. Hindawi
Jučaitė A (2002) Dopaminergic modulation of cerebral activity and cognitive functions. Medicina
38:357–362
Kandratavicius L et al (2012) Psychiatric comorbidities in temporal lobe epilepsy: possible
relationships between psychotic disorders and involvement of limbic circuits. Braz J Psychiatry
34(4):454–466. SciELO Brasil
Kapur S, Mamo D (2003) Half a century of antipsychotics and still a central role for dopamine D2
receptors. Prog Neuropsychopharmacol Biol Psychiatry 27(7):1081–1090. Elsevier
Kebabian JW, Calne DB (1979) Multiple receptors for dopamine. Nature 277(5692):93. Nature
Publishing Group
Khan ZU et al (1998) Prominence of the dopamine D2 short isoform in dopaminergic pathways.
Proc Natl Acad Sci U S A 95(13):7731–7736. https://doi.org/10.1073/pnas.95.13.7731. The
National Academy of Sciences
Kimberg DY, D’esposito M, Farah MJ (1997) Effects of bromocriptine on human subjects depend
on working memory capacity. Neuroreport 8(16):3581–3585. LWW
Kochanek PM et al (2015) Emerging therapies in traumatic brain injury. In: Seminars in neurology.
Thieme Medical Publishers, New York, NY, pp 83–100
Kriks S et al (2011) Dopamine neurons derived from human ES cells efficiently engraft in animal
models of Parkinson’s disease. Nature 480(7378):547. Nature Publishing Group
Lacey MG, Mercuri NB, North RA (1987) Dopamine acts on D2 receptors to increase potassium
conductance in neurones of the rat substantia nigra zona compacta. J Physiol 392(1):397–416.
Wiley Online Library
Lalley PM (2009) D1/D2-dopamine receptor agonist dihydrexidine stimulates inspiratory motor
output and depresses medullary expiratory neurons. Am J Physiol Regul Integr Comp Physiol
296(6):R1829–R1836. American Physiological Society
Laruelle M (2014) Schizophrenia: from dopaminergic to glutamatergic interventions. Curr Opin
Pharmacol 14:97–102. Elsevier
Laruelle M et al (1996) Single photon emission computerized tomography imaging of
amphetamine-induced dopamine release in drug-free schizophrenic subjects. Proc Natl Acad
Sci U S A 93(17):9235–9240. National Acad Sciences
180 S. Kaur et al.
Levite M (2016) Dopamine and T cells: dopamine receptors and potent effects on T cells, dopamine
production in T cells, and abnormalities in the dopaminergic system in T cells in autoimmune,
neurological and psychiatric diseases. Acta Physiol. 216(1):42–89. https://doi.org/10.1111/
apha.12476. John Wiley & Sons, Ltd (10.1111)
Lidow MS et al (1991) Distribution of dopaminergic receptors in the primate cerebral cortex:
quantitative autoradiographic analysis using [3H] raclopride,[3H] spiperone and
[3H] SCH23390. Neuroscience 40(3):657–671. Elsevier
Loma I, Heyman R (2011) Multiple sclerosis: pathogenesis and treatment. Curr Neuropharmacol 9
(3):409–416. Bentham Science Publishers
Lud Cadet J, Jayanthi S, McCoy MT, Beauvais G, Cai NS (2010) Dopamine D1 receptors,
regulation of gene expression in the brain, and neurodegeneration. CNS Neurol Disord Drug
Targets 9(5):526–538
Lv C et al (2018) Dopamine D2-like receptors (DRD2 and DRD4) in chickens: tissue distribution,
functional analysis, and their involvement in dopamine inhibition of pituitary prolactin expres-
sion. Gene 651:33–43. Elsevier
Madikians A, Giza CC (2006) A clinician’s guide to the pathophysiology of traumatic brain injury.
Indian J Neurotrauma 3(01):9–17. Thieme Medical and Scientific Publishers Private Ltd.
Maiti P, Manna J, Dunbar GL (2017) Current understanding of the molecular mechanisms in
Parkinson’s disease: targets for potential treatments. Transl Neurodegener 6:28. https://doi.org/
10.1186/s40035-017-0099-z. England
Malo M et al (2012) Investigation of D1 receptor–agonist interactions and D1/D2 agonist selectivity
using a combination of pharmacophore and receptor homology modeling. Chem Med Chem 7
(3):483–494. Wiley Online Library
Martin EI et al (2009) The neurobiology of anxiety disorders: brain imaging, genetics, and
psychoneuroendocrinology. Psychiatr Clin North Am 32(3):549–575. https://doi.org/10.1016/
j.psc.2009.05.004. United States
Martorana A, Koch G (2014) Is dopamine involved in Alzheimer’s disease? Front Aging Neurosci
6:252. Frontiers
Meiser J, Weindl D, Hiller K (2013) Complexity of dopamine metabolism. Cell Commun Signal 11
(1):34. BioMed Central
Mendez I et al (2008) Dopamine neurons implanted into people with Parkinson’s disease survive
without pathology for 14 years. Nat Med 14(5):507. Nature Publishing Group
Mishra A, Singh S, Shukla S (2018) Physiological and functional basis of dopamine receptors and
their role in neurogenesis: possible implication for Parkinson’s disease. J Exp Neurosci
12:1179069518779829
Nagatsu T, Levitt M, Udenfriend S (1964) Tyrosine hydroxylase the initial step in norepinephrine
biosynthesis. J Biol Chem 239(9):2910–2917. American Society for Biochemistry and Molec-
ular Biology
Nobili A et al (2017) Dopamine neuronal loss contributes to memory and reward dysfunction in a
model of Alzheimer’s disease. Nat Commun 8:14727. Nature Publishing Group
Nyberg L et al (2016) Dopamine D2 receptor availability is linked to hippocampal–caudate
functional connectivity and episodic memory. Proc Natl Acad Sci 113(28):7918–7923. National
Acad Sciences
Onali P, Olianas MC, Bunse B (1988) Evidence that adenosine A2 and dopamine autoreceptors
antagonistically regulate tyrosine hydroxylase activity in rat striatal synaptosomes. Brain Res
456(2):302–309. Elsevier
Paladini CA, Roeper J (2014) Generating bursts (and pauses) in the dopamine midbrain neurons.
Neuroscience 282:109–121. Elsevier
Paredes VEB et al (2015) Reduced D2/D3 receptor binding of extrastriatal and striatal regions in
temporal lobe epilepsy. PLoS One 10(11):e0141098. Public Library of Science
Patel KR et al (2014) Schizophrenia: overview and treatment options. Pharm Ther 39(9):638.
MediMedia, USA
5 Pharmacology of Dopamine and Its Receptors 181
Abstract
Keywords
Serotonin · 5-hydroxytryptamine (5HT) · Serotonin receptors · SSRIs
Abbreviations
5-CT 5-Carboxamidotryptamine
5-HIAA 5-Hydroxyindoleacetic acid
6.1 Introduction
Serotonin is a chemical messenger that is available in each tissue of the human body
as well as identified in plants and aerobic organisms like bacteria. Serotonin is a
ubiquitous monoamine acting as a neurotransmitter and hormone (Mohammad-
Zadeh et al. 2008), which is also recognized as 5-hydroxytryptamine (5-HT) (Shad
2017). Serotonin was first independently recognized in the late 1940s in the USA
and Italy. At that time it was known as “Serotonin” in the USA and “Enteramine” in
Italy. In 1950, it was confirmed that the structure of both the compounds were same.
In the mid-1950s, serotonin was identified in the brain of animals (Glennon and
6 Pharmacology of Serotonin and Its Receptors 185
NH2
CH2 C COOH H2 NH2 H2 H2
H HO C CH COOH C C NH2
HO
N Tryptophan
H N Decarboxylase N
hydroxylase H H
Tryptophan 5-Hydroxytryptophan
Serotonin (5-Hydroxytryptamine)
MAO+Aldeh
ydrogenase
H2 O
HO C C OH
N
H
5-Hydroxyindoleacetic acid
Fig. 6.1 Synthesis and degradation of serotonin (5-hydroxytryptamine) (Sources: Pytliak et al.
2011)
Dukat 2002). It is also found in the platelets, neurons, and GIT (enterochromaffin
cells of the GIT mucosa). Over 90% (Shajib and Khan 2015) of the total amount in
the body is present in the enterochromaffin cells of the gut and a lesser amount in the
brain and retina (Pytliak et al. 2011; Peterlin and Rapoport 2007). In the brain, cell
bodies of serotonergic neurons are mostly located in the brainstem midline with
broad axonal ridge to all areas of the CNS (Peterlin and Rapoport 2007). Serotonin is
associated with various diseases like schizophrenia, anxiety, depression, hyperten-
sion, migraine, carcinoid diarrhea, vomiting, irritable bowel syndrome, pulmonary
hypertension, and eating disorders (Pauwels 2003; De Ponti 2004).
Serotonin secreted by the nuclei in the median raphe of CNS is transferred toward
the spinal cord and other parts of the brain including the hypothalamus. Serotonin
discharged from the enterochromaffin cells of GIT finds it own way out of the tissues
into the blood, where it is taken up by the blood platelets (Zarrindast et al. 2014).
Serotonin synthesis depends on two factors: (1) accessibility of tryptophan which is an
amino acid obtained on or after nutritional ingestion, and (2) the action of rate-limiting
enzymes tryptophan hydroxylase (Peterlin and Rapoport 2007). Serotonin synthesis
starts through active uptake of tryptophan in neurons and enterochromaffin cell by use
of amino acid transporters (Pytliak et al. 2011; Upadhyay 2003). Initially, in the
presence of tryptophan hydroxylase, tryptophan is converted into
5-hydroxytryptophan, which is finally converted into 5-hydroxytryptamine (serotonin)
with the help of aromatic L-amino acid decarboxylase (Fig. 6.1) (Berger et al. 2009).
Serotonin is mainly degraded through oxidative deamination, catalyzed by mono-
amine oxidase A (MAO A), and followed by oxidation to 5-hydroxyindoleacetic acid
(5-HIAA) (Ni and Watts 2006). In the brain, it is protected from enzymatic degrada-
tion by storing in synaptic vesicles of neurons. When 5-HT is released from the storage
vesicles, it is either metabolized into 5-HIAA or reuptaken into the presynaptic
186 S. Deka et al.
MAO
Release
Reuptake
Postsynaptic Neurons
neurons by the serotonin transporter (5-HTT or SERT) (Fig. 6.2) (Hamel and Currents
2007).
Serotonin possesses trophic factors in the early stages of pregnancy in humans.
Serotonin secreted in mother’s enterochromaffin cells of the GIT moves to the
platelets in mother’s blood. In the meantime from early stage of pregnancy, the
fetus additionally begins its own 5-HT secretion process in the nuclei of the
midbrain. After distribution of 5-HT by serotonergic neurons through the body
and brain of the fetus, it expands division, relocation, and development of peripheral
and central tissues (Shad 2017). Serotonin has vast numbers of physiological activity
like modulation of platelet aggregation, contraction of vascular and nonvascular
smooth muscle, regulation of appetite, mood, anxiety, controlling of body tempera-
ture, wakefulness, perception of sexual behavior, and hormone secretion (Upadhyay
2003; Kroeze et al. 2002). Serotonin and its receptors play a significant role in the
functioning of the brain and therefore, dysregulation of serotonin system leads to
various psychiatric and neurological disorders (Berger et al. 2009).
Serotonin, a notable neurotransmitter in the CNS, also plays a significant role in
peripheral tissues as well as in immunity. The growing human body is the proof of
recommending that a wide range of immune cells express the mechanism to create,
store, react, and transport serotonin, including mast cells, T cells, macrophages,
platelets, and dendritic cells. Moreover, there is rising evidence of a possible
association between mood disorder, serotonin, and T cells. However, it is not clear
how serotonin associates with immunity (Wu et al. 2019).
6 Pharmacology of Serotonin and Its Receptors 187
6.2.2 GIT
The direct action of serotonin on 5-HT2 smooth muscle receptors of GIT causes
enhancing tone and facilitates peristalsis movement. In the enteric nervous system,
activation of 5-HT4 receptors causes increased release of acetylcholine. Serotonin
inhibits gastric secretion but increases mucous production. Overproduction of sero-
tonin in carcinoid tumors causes severe diarrhea (Katzung 2018). Serotonin is also
involved in digestion right after food enters the body. Actuation of taste bud cells on
the tongue causes serotonin release onto sensory afferent nerves that move the taste
information to the CNS. When food enters in the GIT, serotonin regulates the
peristaltic movement and secretions. Modified 5-HT signaling has been concerned
in bowel disorders like irritable bowel syndrome (IBS) (Gershon and Tack 2007) and
is efficiently treated by the medication targeting both 5-HT3 and 5-HT4 receptors.
Excessive serotonin secretion in GIT can also activate the 5-HT3 receptors present in
afferent vagal nerves that innervate vomiting center present in the brainstem, which
also justifies why 5-HT3 antagonists like ondansetron are valuable antiemetic
(Berger et al. 2009).
Serotonin shows a significant feature in the development of the brain (Nordquist and
Oreland 2010). In the CNS, serotonergic neurons secrete serotonin which influences
various functions like temperature regulation, appetite, mood, hormone secretion,
cognition, sensory perception, and motor activity (Shajib and Khan 2015). A
decrease in serotonin levels leads to various psychiatric disorders like depression,
suicidal tendency, and violence. Notably, females are more prone to depressive
disorders than males, because the rate of serotonin synthesis is only about half of
188 S. Deka et al.
that in males. Depressive disorders are prominent in aged person which may
be because of less serotonin synthesis (Sibley et al. 2018).
6.2.5 Platelets
In platelets, the synthesis of serotonin does not occur but expresses the mechanisms
for serotonin uptake, storage, and exocytotic release. Serotonin promotes platelet
aggregation by binding to the platelet 5HT2A receptors. Serotonin released from
adherent platelets causes vasodilation, when the endothelium is undamaged, aiding
proper blood flow, whereas in damaged endothelium serotonin causes constriction
and further impairs the blood flow. These impacts of platelet-derived 5-HT are
believed to be significant in vascular disease (Sibley et al. 2018; Rang et al. 2016).
6.3.1 Neurotransmitter
Serotonin
Syndrome Obesity Depression
Anxiety Schizophrenia
ROLE OF SEROTONIN
Migraine
GI Disorder
6.3.5 Cancer
Recently it has been found that serotonin plays an important role in human tumor
cells of different origins like glioma, carcinoid, and carcinomas. Serotonin is
involved in the migration of cancer cells, tumor angiogenesis, and metastatic
dissemination. Different studies show that the levels of serotonin play a major role
in cancer cell growth (Sarrouilhe and Mesnil 2019).
Serotonin has an important role in normal and dysfunctional GIT motility. Serotonin
receptors especially 5-HT3 and 5-HT4 receptors are targeted to treat various GIT
motility disorders. Different studies have been performed on rat to investigate
the relevance of serotonin with colonic motility (Kendig and Grider 2015). Serotonin
is a significant flagging particle in the gut particularly in enterocytes, smooth
muscles, and enteric neurons. Most of the body serotonin is available in enterochro-
maffin cells. Serotonin promotes the activation of extrinsic and intrinsic afferent
neurons to start the peristaltic and secretary reflexes and convey information to the
CNS. Serotonin is reuptaken by the serotonin transporter (SERT) in the enterocytes
or neurons. Exogenous serotonin application inspires such a large number of
responses that it is hard to figure out which is physiologically relevant. This impact
is to a great extent because of the nearness of multiple receptor subtypes, which
seem, by all accounts, to be available on a few classes of myenteric neurons, on
smooth muscle cells, and on epithelial cells. IBS is an unpredictable issue that is
associated with altered gastrointestinal motility, discharge, and sensation. Changed
serotonin signaling may prompt both intestinal and extraintestinal frameworks in
IBS (Sikander et al. 2009).
6.3.7 Obesity
Serotonin receptors present in the CNS are related with the parameter of food
ingestion leading to obesity (Thomsen et al. 2008). The capacity to store and prepare
vitality is essential for physiologic capacity. Overabundance vitality is put away in
fat tissue as triglycerides, which is discharged as free unsaturated fats when required,
through cell forms firmly managed by insulin. Fat cell functioning is required to
regulate the metabolism of lipid and body glucose level. Studies have been conducted
to understand the effect of 5-HT on lipid digestion and glucose homeostasis and
cytokine emission (Fex and Stenkula 2019). Obesity increases the risk of various
diseases like diabetes mellitus, congestive heart failure, coronary heart diseases,
stroke, hypertension, and osteoarthritis (Smith et al. 2009).
6 Pharmacology of Serotonin and Its Receptors 191
6.3.8 Migraine
Migraine pain is the most continuous neurological issue in the growing population
around the world, influencing up to 12% of the all inclusive community and more
common in women about 25%. It highly affects the general public because of its
crippling nature and in that, decreased personal satisfaction and expanded absence
from work. Cerebral pain is the essential clinical sign and it has been related with
hereditary affectability of neurovascular responses to specific improvements or to
cyclic changes in the central nervous system. Among the numerous synapses in the
brain, the serotonergic system (5-HT) from the brainstem raphe core has been most
convincingly ensnared in migraine pathophysiology. The changes in metabolism of
serotonin and serotonin-mediated responses during the migraine pain recommend
that migraine pain is a result of a central neurochemical imbalance that includes a
low serotonergic character, although the correct flow between serotonergic neuro-
transmission to the sign and symptoms of migraine pain is still not completely
understood (Hamel and Currents 2007).
Gaddum and Picarelli proposed the arrangement of 5-HT receptors in 1957, when it
was exhibited that practical reactions of the guinea pig ileum could be mostly
obstructed by morphine (M); at the same time the rest of the reaction can be
obstructed by dibenzyline (D) and named them as M and D receptors, respectively
(Göthert 2013).
Presently serotonin receptor families are classified into seven families (Table 6.1)
naming 5-HT1 to 5-HT6. All classes are G-protein-coupled receptors (GPCRs)
5-HT
5-HT4F
5-HT4G
5-HT4H
except 5-HT3 receptor which is a ligand-gated ion channel (Kroeze et al. 2002).
Based on sequence and pharmacological activity, all 5-HT receptors are further
subdivided (Fig. 6.4) through alternative splicing, RNA editing, etc. (Barnes and
Sharp 1999).
The 5-HT1 receptor is the largest class of 5-HT receptor, which has five subtypes
5-HT1A, 5-HT1B, 5-HT1D, 5-HT1E, and 5-HT1F (Fig. 6.4). All the subfamilies of
5-HT1 receptor show high affinity for the synthetic agonist,
5-carboxamidotryptamine (5-CT), except 5-HT1E and 5-HT1F receptors (Lanfumey
and Hamon 2004). The mechanism of these receptors is to decrease adenylyl cyclase
(AC) and cAMP levels by pairing to Gi/o protein. Additionally signal transduction
mechanisms also have been described (Pytliak et al. 2011).
The 5-HT3 receptor is only 5-HT receptor which is a part of ionotropic ligand-gated
ion channel. The structure and functions of 5-HT3 receptors resemble the members
of the cys-loop ligand-gated ion channel family (Thompson and Lummis 2007). The
receptors are recognized on neurons of both central and peripheral origin, where they
produce quick depolarization because of a transient internal current, resulting in the
opening of nonselective cation channels (Na+, Ca++ influx, K+ efflux). 5-HT3
receptors cause vomiting and nausea during radiotherapy and chemotherapy process,
which are treated with 5-HT3 antagonist like ondansetron, granisetron, tropisetron,
palonosetron, and dolasetron (Table 6.2) (Hoyer et al. 2002; De Ponti 2004). At
196 S. Deka et al.
These receptors have two subfamilies, i.e., 5-HT5A and 5-HT5B (Fig. 6.4). They are
coupled to Gi/o to inhibit AC. Humans only express functional 5-HT5A receptors.
From their regions, it has been theorized that they might be engaged with feeding,
anxiety, motor control, depression, adaptive behavior, learning, memory consolida-
tion, and brain expansion (Thomas 2006). Disturbance of 5-HT neuron-glial
associations might be engaged with the advancement of certain CNS pathologies
including Alzheimer’s disease, Down’s syndrome, and some drug-actuated
6 Pharmacology of Serotonin and Its Receptors 197
development deficits. After analyzing the location of chromosome, it was found that
for human 5-HT5A receptors: position 7q36 and chromosome 7; 5-HT5B receptors:
position 2q11–13, chromosome 2. 5-HT5A consists of 357 amino acids but in
5-HT5B receptor, end codons are present in the gene. Studies on 5-HT5A and
5-HT5B are very less among the other serotonin receptors (Nelson 2004).
It is the first receptor of 5-HT which is coupled to AC (Woolley et al. 2004) and
increases cAMP intracellular level by pairing to Gs protein. 5-HT6 receptors are
located predominantly inside limbic and extrapyramidal cerebral zones in the CNS,
which suggest that it is important for motor control and cognition (Sibley et al.
2018). The accurate scientific importance of 5-HT6 receptors remains still indistin-
guishable. 5-HT6 receptors have a role in learning and memory process. It also plays
a role in obesity. Additionally, the 5-HT6 receptors have been recommended to be
concerned in psychotic disease such as epilepsy and anxiety (Kitson 2007). The
5-HT6 receptor was first discovered in rats and humans in 1993 and 1996, respec-
tively with the help of molecular biology. The 5-HT6 receptor shows a unique
pharmacological activity of high affinity toward antipsychotic molecules and tricy-
clic and atypical antidepressant molecule like clozapine, loxapine, amitriptyline, and
mianserin. In 1998, first selective 5-HT6 receptor antagonist was reported in humans,
i.e., Ro04–6790; chemically it is 4-amino-N-(2,6-bis-methyl-amino-pyrimidin-4-
yl)-benzene sulphonamide. Ro04–6790 has less penetration through BBB, but
when 30 mg/kg dose intraperitoneal was given it showed 70% receptor occupancy
from CSF levels (Woolley et al. 2004).
Azapirones like buspirone, gepirone, and ipsapirone are agonists of 5-HT1A receptor
which is partially active. These are used as anxiolytic drugs and may work on the
autoreceptors to reduce serotonergic activity (Katzung 2018).
Dexfenfluramine (Fig. 6.5) is another agonist of 5-HT receptors, which is a
selective agonist. It may be broadly used as a hunger suppresser but it was with-
drawn due to cardiac valvulopathy (Katzung 2018).
Lorcaserin (Fig. 6.5) is approved for weight loss medication which is a 5-HT2C
agonist (Fig. 6.7) (Katzung 2018). The drug is thought to reduce food consumption
and increase satiety by selectively activating 5HT2C receptors on hypothalamus.
Chemically lorcaserin is 1R-8-chloro-2,3,4,5-tetrahydro-1-methyl-1H-3-benzapine
(Smith et al. 2009). In humans, lorcaserin shows 100-fold selectivity for 5-HT2C
versus the closely related 5-HT2B receptor and 17-fold selectivity over the 5-HT2A
receptor (Thomsen et al. 2008; Smith et al. 2008). There is no useful action at
additional 5-HT receptors when the concentration of lorcaserin is less than
1 micromol per liter. This leads to the minimal adverse effects than that of nonselec-
tive 5-HT receptor agonist (Smith et al. 2009). The clinical dose recommendation for
18 years and older patient of lorcaserin is 10 mg, two times in a day. On oral
administration, lorcaserin absorbed rapidly greater than 90%. The Cmax is about
1.5–2 h and t1/2 is about 11 h. About 70% drugs are bound to plasma protein.
Lorcaserin is metabolized in the liver and the main metabolite is lorcaserin
sulfamate. It is excreted by the kidney through urine and the main metabolite is N-
carbamoylglucuronides (Bai and Wang 2011). Common adverse effects are
vomiting, nausea, diarrhea, constipation, fatigue, UTI, upper respiratory tract
infections, rashes, headache, dizziness and back pain; memory and attention defi-
ciency are also seen in 1.9% of patients (Brashier et al. 2014).
The 8-hydroxydipropylamino tetraline (8-OH DPAT) is an aminotetralin deriva-
tive and highly selective 5-HT1A agonist. It is used as an experimental tool and not
used therapeutically because of its low oral bioavailability (Tripathi 2013).
Sumatriptan and other triptans are selective 5-HT1B/1D agonist and effective to
treat acute migraine pain attacks (Fig. 6.7). Migraine pain is a disease characterized
by pulsating pain in the head lasting for 4–8 h and often associated with vomiting,
nausea, sensitivity to sound and light, flashes of light, and other symptoms. Dilata-
tion of certain cranial vessels causes migraine pain; selective 5-HT1B/1D agonists
(triptans) constrict the dilated cerebral blood vessels. Triptans have three
mechanisms of action which relieve the migraine pain. The mechanisms are by
direct effect on vascular smooth cells which causes vasoconstriction of cranial
vessels, reduction of nociceptive neurotransmission, and reduction of discharge of
vasoactive neuropeptides (Tepper et al. 2002).
In 1984, Sumatriptan (Fig. 6.5) was discovered. It is the first selective 5-HT1B/1D
agonist among the triptans. At first, it was introduced as injectable, followed by
tablet, nasal spray, and suppositories (Dahlöf 2001). Sumatriptan demonstrates its
6 Pharmacology of Serotonin and Its Receptors 199
N CH3
NH H2 CH3
O H2 H2 N N CH2 CH3
C C N OH H2C O S O H2C C N
C C H2C CH3
N N CH2
H2 H2 CH2 CH3
O N
H
Buspirone 8-Hydroxy-DPAT
Sumatriptan
N CH3
CH3
H3C CH3 N NH2
N H2 HC CH CH3
H2 H2C CH2 H2 CH2 C H2C
C O C H2C H3CO
S CH2 O S O N
NH O N H
O N H NH OCH3
H
CH3 I
O
Eletriptan Naratriptan DOI
Zolmitriptan
NH2
H2C
CH2
HO N R
NH CH3
Cl N N N
H
H3C
H3C
N CH3
O
O N
H Cl
N N
N N H
N N H2N O
H
CF3
Rizatriptan Prucalopride
Dexfenfluramine
Fig. 6.5 5-HT receptor agonists (Sources: Glennon and Dukat 2002)
The bioavailability of sumatriptan is about 15% in intranasal, 14% after oral, and
96% after subcutaneous administrations. Incomplete absorption and presystemic
metabolism cause low bioavailability. The therapeutic dose is 8–66 ng/ml. After
subcutaneous administration Cmax is 25 min and after oral or intranasal administra-
tion the Cmax is 60–90 min. The t1/2 is 1.5–2.6 h (Femenia-Font et al. 2005). Adverse
effects of sumatriptan are difficulty and abnormal thinking, tiredness, dizziness,
agitation, fatigue, tremor, vertigo, etc. (Dodick and Martin 2004).
Zolmitriptan (Fig. 6.5) is another Triptans which have higher oral bioavailability
than sumatriptan, about 40% (Tepper et al. 2002). Chemically zolmitriptan is (S)4-
[3-[2-dimethylamino-ethyl]-1H-indol-5-yl]methyl-2-oxazolidinone (Goads and
Boes 2001). It is a selective 5-HT1B/1D receptor agonist. It is used effectively in
the treatment of migraine and adolescent migraine (Lewis et al. 2007). It is also used
in the treatment of cluster headache (Bahra et al. 2000). The t1/2 is 3 h and Tmax is 2 h.
Zolmitriptan is metabolized in the liver and eliminated with cytochrome P450
pathways (Goads and Boes 2001).
Naratriptan (Fig. 6.5) has 60% more bioavailability than sumatriptan, greater
lipophilicity, less readily metabolized, and better CNS penetration (Tepper et al.
2002). It belongs to triptans, a second-generation antimigraine drug. Naratriptan is
5-HT1B/1D receptor agonists. Chemically naratriptan is (N-methyl-3-1-methyl-4-
piperidinyl)-1-H-indole-5-ethanesulphonamide hydrochloride. Among the orally
administered triptans, naratriptans have highest bioavailability. The approximate
dose is 35 microgram per Kg. The pharmacokinetics of naratriptan is that it has
longest t1/2 of about 5–6 h after oral administration. 70% of naratriptan eliminates
unchanged; only a considerable part is subjected to P450 metabolism. Naratriptan
has fewer side effects (Lambert 2005).
Rizatriptan (Fig. 6.5) has high bioavailability than sumatriptan, is more potent,
and has a rapid onset of action (Tepper et al. 2002). Chemically rizatriptan is N,N-
dimethyl-2-[5-(1H-1,2,4-triazol-1-ylmethyl)-1H-indol-3-yl]ethanamine. Metabo-
lism of rizatriptan occurs in the liver by MAO-A. Rizatriptan-N-oxide and
triazolomethyl indole-3-acetic acid are two major metabolites of rizatriptan; both
are inactive. N-monodesmethyl-rizatriptan is a minor active metabolite of rizatriptan.
After oral administration, bioavailability is 45%, Cmax is 1–1.5 h, t1/2 is about 2–3 h,
and 14% bound to plasma protein. In 1998, the FDA approved rizatriptan and it is
available as oral disintegrating tablets in a dose of 5 mg or 10 mg (Wellington and
Jarvis 2002; Kacperski and O’brien 2012). Rizatriptan is used in the migraine with
aura and migraine without aura treatment in adult patients (Wellington and Jarvis
2002).
Cisapride (Fig. 6.5) is a benzamide derivative (Crowell 2004) and it agonizes the
5-HT4, which is used to treat GIT disorders like gastroesophageal motility and reflux
(Fig. 6.7) (Katzung 2018). Cisapride speeds up the process of stomach emptying and
intestinal passage but it has limitations on varying bowel functions. Some studies
confirmed that it is used in chronic constipation. But nowadays it is available only
for considerate use in the USA because of its toxicity like patient deaths and cardiac
dysrhythmias (Crowell 2004).
Tegaserod (Fig. 6.5) is a partial agonist highly selective for the 5-HT4 receptor
(Camilleri 2001), which is used for IBS and constipation. It is an
6 Pharmacology of Serotonin and Its Receptors 201
These are a variety of compounds, which block serotonergic receptors and antago-
nize the action. Some newly developed antagonists are described below:
Cyproheptadine (Fig. 6.6) has potent 5-HT2 blocking action. The major clinical
uses include management of the smooth muscle manifestations, carcinoid tumor, and
cold-induced urticaria. It is used in children for increasing appetite and poor eater for
weight gaining. Cyproheptadine also reduces muscle spasms following spinal cord
(Hoyer et al. 2002) injury, in which constitutive activity of 5-HT2C receptors is
connected with rising Ca2+ currents leading to spasms. It has some side effects like
dry mouth, drowsiness, weight gain, and confusion ataxia (Tripathi 2013).
Methysergide (Fig. 6.6) antagonizes some action of 5-HT on smooth muscles
including blood vessels. It is a 5-HT2A/2C antagonist. It is used in migraine prophy-
laxis and in the treatment of carcinoid and postgastrectomy dumping syndrome.
Long-term use causes abdominal, pulmonary, and endocardial fibrosis (Tripathi
2013). Clinically, methysergide is available in tablet form of 1 mg. Long-term
uses of methysergide has adverse effects like pleura pulmonary fibrosis, fibrotic
thickening, and retroperitoneal fibrosis (Dahlöf and Maassen Van Den Brink 2012).
Ketanserin (Fig. 6.6) is a 5-HT2 receptor antagonist. Among 5-HT2 receptors,
ketanserin shows stronger blockade in 5-HT2A than 5-HT2C. It antagonizes
202 S. Deka et al.
CH3
O N
F CH3
CH3
NH HO
NH C CH3 N
N N H
N CH3 N
O O
OCH3
O
N
WAY100135 Spiperone GR-55562
O
HN S CH3
O O
H2 H2 O
O N
O N C C N C F
N
H
N
CH3
Ketanserin
GR113808
H
N
N N
O H2C
Cl N
H3C N
N CH3
N
N
CH3
Clozapine Cyproheptadine
Ondansetron
CH3
NH
HO CH3
O N
H
N
CH3
Methysergide
Fig. 6.6 5-HT receptor antagonists (Sources: Glennon and Dukat 2002)
Drug Example
Sumatriptan, Rizatriptan,
Zolmitriptan, Naratriptan 5-HT1D Migraine
Chemotherapy induced
Ondansetron, Tropisetron 5-HT3 emesis
Cisapride, Mosapride,
Renzapride 5-HT4 Gastrointestinal disorder
Extracellular
Intracellular
NH2 COOH
H
H N
O N
CH3 H2 H2 H2 H2 O
F3C C C C C C O CH3 CH2
F3C O
N H H O
2 2
O C C NH2
Fluoxetine Fluvoxamine Paroxetine
F F
CH3
CH3 N
Cl N CH3 N
H O
Cl NC
CH3
N
Sertraline Citalopram Imipramine NH2
NH
CH3
Desipramine
Fig. 6.9 Structure of some SERT inhibitors (Sources: Glennon and Dukat 2002)
absorbed from GIT and metabolized in the liver. Complete absorption takes place
from the GIT in case of sertraline. The Cmax of sertraline is 6–8 h (Hiemke and
HÄrtter 2000).
symptoms, and the elimination of extra situation (Birmes et al. 2003). On clinical
ground severe toxicity is diagnosed and characterized by a quickly rising tempera-
ture and inflexibility. Mild toxicity is difficult to recognize from numerous ailments
or other unfriendly medication impacts (Buckley et al. 2014). Sternbach criteria are
used for the analysis of serotonin syndrome, but it is still complicated in cases of
benign symptoms or normal neurological test results (Birmes et al. 2003). Serotonin
syndrome or toxicity is treated primarily by caring, charcoal lavage, consisting of
external cooling with blankets, and dialysis in the case of lithium overdose (Ener
et al. 2003). Generally, discontinuation of serotonergic medications is done to
overcome from toxicity (Buckley et al. 2014). Benzodiazepines are used in some
neurological symptoms, including serious myoclonus and hyperreflexia. In severe
toxicity cases cyproheptadine is recommended (Frank 2008).
of ondansetron (5-HT3 antagonist) on the quantity of water in large and small bowel.
The study will measure the mode of action of ondansetron in lactulose-induced
diarrhea. The study was initiated in October 2018 and estimated to be completed by
August 2019 (ClinicalTrials.gov Identifier: NCT03833999).
Aprepitant in preventing nausea and vomiting: A phase II study was performed in
22 participants by Wake Forest University Health Sciences to evaluate the effect of
aprepitant in preventing nausea and vomiting in patients undergoing chemotherapy
and radiation therapy for pancreatic cancer. The drugs used in the study were
aprepitant, gemcitabine hydrochloride, capecitabine, and fluorouracil. The study
was initiated in August 2006 and completed in August 2012 (ClinicalTrials.gov
Identifier: NCT01534637).
Granisetron and myofascial pain: A randomized phase IV study was performed
in 40 participants by Karolinska Institute to evaluate the effect of granisetron on
myofascial pain in the orofacial muscles. The study was initiated in March 2007 and
completed in July 2015 (ClinicalTrials.gov Identifier: NCT02230371).
ONZETRA® Xsail® and episodic migraine: A randomized phase III study was
undertaken in 420 participants by Avanir pharmaceuticals to evaluate the safety and
efficacy of ONZETRA® Xsail® (sumatriptan nasal powder) for the acute treatment
of episodic migraine with or without aura in adolescents. The study was designed to
examine the efficacy and safety of sumatriptan nasal powder for treating migraine in
12–17 years old patients. The study was initiated in November 2017 and completed
by November 2019 (ClinicalTrials.gov Identifier: NCT03338920).
Ondansetron and CNS distribution: A non-randomized phase I study has been
performed in 18 participants by Washington University School of Medicine to
determine the time course of plasma and CSF concentrations of IV ondansetron in
healthy subjects, with and without selective inhibition of Pgp efflux transporter. The
study was initiated in May 2019 and estimated to be completed by June 2020
(ClinicalTrials.gov Identifier: NCT03809234).
Ondansetron and hypotension: A randomized phase IV study has been performed
in 100 participants by Hospital de base to verify the hypothesis that ondansetron IV
(5-HT3 receptor antagonist) decreases the occurrence of hypotension induced by
spinal anesthesia. The study was initiated in March 2019 and estimated to be
completed by March 2020 (ClinicalTrials.gov Identifier: NCT03973411).
6.10 Conclusion
References
Bahra A, Gawel M, Hardebo J-E, Millson D, Breen S, Goadsby P (2000) Oral zolmitriptan is
effective in the acute treatment of cluster headache. Neurology 54:1832–1839
Bai B, Wang Y (2011) The use of lorcaserin in the management of obesity: a critical appraisal. Drug
Des Devel Ther 5:1
Barnes NM, Neumaier JF (2011) Neuronal 5-HT receptors and SERT. Tocris Biosci Sci Rev Ser
34:1–16
Barnes NM, Sharp T (1999) A review of central 5-HT receptors and their function. Neuropharma-
cology 38:1083–1152
Beecher K, Belmer A, Bartlett SE (2019) Anatomy of the serotonin transporter. Serotonin. Elsevier,
Amsterdam
Berger M, Gray JA, Roth BL (2009) The expanded biology of serotonin. Annu Rev Med
60:355–366
Birmes P, Coppin D, Schmitt L, Lauque D (2003) Serotonin syndrome: a brief review. CMAJ
168:1439–1442
Bortolozzi A, Díaz-Mataix L, Scorza MC, Celada P, Artigas F (2005) The activation of 5-HT2A
receptors in prefrontal cortex enhances dopaminergic activity. J Neurochem 95:1597–1607
Brashier DB, Sharma A, Dahiya N, Singh S, Khadka A (2014) Lorcaserin: a novel antiobesity drug.
J Pharmacol Pharmacother 5:175–178
Briejer MR, Bosmans J-P, Van Daele P, Jurzak M, Heylen L, Leysen JE, Prins NH, Schuurkes JA
(2001) The in vitro pharmacological profile of prucalopride, a novel enterokinetic compound.
Eur J Pharmacol 423:71–83
Buckley NA, Dawson AH, Isbister GK (2014) Serotonin syndrome. BMJ 348:g1626
Camilleri M (2001) tegaserod. Aliment Pharmacol Ther 15:277–289
Cazzola M, Matera MG (2000) 5-HT modifiers as a potential treatment of asthma. Trends
Pharmacol Sci 21:13–16
Ciranna Á (2006) Serotonin as a modulator of glutamate-and GABA-mediated neurotransmission:
implications in physiological functions and in pathology. Curr Neuropharmacol 4:101–114
Crowell MD (2004) Role of serotonin in the pathophysiology of the irritable bowel syndrome. Br J
Pharmacol 141:1285–1293
Culy CR, Bhana N, Plosker GL (2001) Ondansetron. Paediatr Drugs 3:441–479
Dahlöf CG (2001) Sumatriptan: pharmacological basis and clinical results. Curr Med Res Opin 17:
s35–s45
Dahlöf C, Maassen Van Den Brink A (2012) Dihydroergotamine, ergotamine, methysergide and
sumatriptan–basic science in relation to migraine treatment. Headache 52:707–714
De Ponti F (2004) Pharmacology of serotonin: what a clinician should know. Gut 53:1520–1535
Dodick DW, Martin V (2004) Triptans and CNS side-effects: pharmacokinetic and metabolic
mechanisms. Cephalalgia 24:417–424
Ener RA, Meglathery SB, Decker WAV, Gallagher RM (2003) Serotonin syndrome and other
serotonergic disorders. Pain Med 4:63–74
Femenia-Font A, Balaguer-Fernandez C, Merino V, Rodilla V, Lopez-Castellano A (2005) Effect
of chemical enhancers on the in vitro percutaneous absorption of sumatriptan succinate. Eur J
Pharm Biopharm 61:50–55
Feng J, Cai X, Zhao J, Yan Z (2001) Serotonin receptors modulate GABAA receptor channels
through activation of anchored protein kinase C in prefrontal cortical neurons. J Neurosci
21:6502–6511
Fex M, Stenkula KG (2019) Serotonin and adipocyte function. Serotonin. Elsevier, Amsterdam
Frampton JE (2009) Prucalopride. Drugs 69:2463–2476
Frank C (2008) Recognition and treatment of serotonin syndrome. Can Fam Physician 54:988–992
Fuseau E, Petricoul O, Moore KH, Barrow A, Ibbotson T (2002) Clinical pharmacokinetics of
intranasal sumatriptan. Clin Pharmacokinet 41:801–811
210 S. Deka et al.
Galligan J (2002) Ligand-gated ion channels in the enteric nervous system. Neurogastroenterol
Motil 14:611–623
Gershon MD, Tack J (2007) The serotonin signaling system: from basic understanding to drug
development for functional GI disorders. Gastroenterology 132:397–414
Glennon RA, Dukat M (2002) Serotonin receptors and drugs affecting serotonergic neurotransmis-
sion. In: Foye’s Textbook of Medicinal Chemistry. Lippincott Williams & Wilkins,
Philadelphia, pp 315–337
Goads PJ, Boes CJ (2001) Zolmitriptan: differences from sumatriptan. Curr Med Res Opin 17:s46–
s50
Göthert M (2013) Serotonin discovery and stepwise disclosure of 5-HT receptor complexity over
four decades. Part I. general background and discovery of serotonin as a basis for 5-HT receptor
identification. Pharmacol Rep 65:771–786
Hamel E, Currents H (2007) Serotonin and migraine: biology and clinical implications. Cephalalgia
27:1293–1300
Hesketh PJ (2008) Chemotherapy-induced nausea and vomiting. N Engl J Med 358:2482–2494
Hiemke C, HÄrtter S (2000) Pharmacokinetics of selective serotonin reuptake inhibitors.
Pharmacol Ther 85:11–28
Hoyer D, Hannon JP, Martin GR (2002) Molecular, pharmacological and functional diversity of
5-HT receptors. Pharmacol Biochem Behav 71:533–554
Kacperski J, O’brien HL (2012) Triptan use in pediatric migraine: focus on rizatriptan. Future
Neurol 7:385–394
Katzung BG (2018) Basic & clinical pharmacology. McGraw Hill Education (India) Private
Limited, New Delhi
Kendig DM, Grider JR (2015) Serotonin and colonic motility. Neurogastroenterol Motil
27:899–905
King MV, Marsden CA, Fone KC (2008) A role for the 5-HT1A, 5-HT4 and 5-HT6 receptors in
learning and memory. Trends Pharmacol Sci 29:482–492
Kitson SL (2007) 5-hydroxytryptamine (5-HT) receptor ligands. Curr Pharm Des 13:2621–2637
Knight AR, Misra A, Quirk K, Benwell K, Revell D, Kennett G, Bickerdike M (2004) Pharmaco-
logical characterisation of the agonist radioligand binding site of 5-HT 2A, 5-HT 2B and 5-HT
2C receptors. Naunyn Schmiedeberg’s Arch Pharmacol 370:114–123
Kroeze WK, Kristiansen K, Roth BL (2002) Molecular biology of serotonin receptors-structure and
function at the molecular level. Curr Top Med Chem 2:507–528
Lambert GA (2005) Preclinical neuropharmacology of naratriptan. CNS Drug Rev 11:289–316
Lanfumey L, Hamon M (2004) 5-HT1 receptors. Curr Drug Targets CNS Neurol Disord 3:1–10
Launay J-M, Herve P, Peoc’H K, Tournois C, Callebert J, Nebigil C, Etienne N, Drouet L,
Humbert M, Simonneau G (2002) Function of the serotonin 5-hydroxytryptamine 2B receptor
in pulmonary hypertension. Nat Med 8:1129–1135
Lewis DW, Winner P, Hershey AD, Wasiewski WW (2007) Efficacy of zolmitriptan nasal spray in
adolescent migraine. Pediatrics 120:390–396
Leysen J (2004) 5-HT2 receptors. Current Drug Targets CNS Neurol Disord 3:11–26
Lummis SC (2012) 5-HT3 receptors. J Biol Chem 287:40239–40245
Maclean MR, Herve P, Eddahibi S, Adnot S (2000) 5-hydroxytryptamine and the pulmonary
circulation: receptors, transporters and relevance to pulmonary arterial hypertension. Br J
Pharmacol 131:161–168
Meneses A (2015) Serotonin, neural markers, and memory. Front Pharmacol 6:143
Mohammad-Zadeh L, Moses L, Gwaltney-Brant S (2008) Serotonin: a review. J Vet Pharmacol
Ther 31:187–199
Müller-Lissner S, Fumagalli I, Bardhan K, Pace F, Pecher E, Nault B, Rüegg P (2001) Tegaserod, a
5-HT4 receptor partial agonist, relieves symptoms in irritable bowel syndrome patients with
abdominal pain, bloating and constipation. Aliment Pharmacol Ther 15:1655–1666
Naughton M, Mulrooney JB, Leonard BE (2000) A review of the role of serotonin receptors in
psychiatric disorders. Hum Psychopharmacol Clin Exp 15:397–415
6 Pharmacology of Serotonin and Its Receptors 211
Thompson AJ, Lummis SC (2007) The 5-HT3 receptor as a therapeutic target. Expert Opin Ther
Targets 11:527–540
Thomsen WJ, Grottick AJ, Menzaghi F, Reyes-Saldana H, Espitia S, Yuskin D, Whelan K,
Martin M, Morgan M, Chen W (2008) Lorcaserin, a novel selective human
5-hydroxytryptamine2C agonist: in vitro and in vivo pharmacological characterization. J
Pharmacol Exp Ther 325:577–587
Tripathi K (2013) Essential of medical pharmacology, Jaypee Brothers Medical Publishers Pvt. Ltd,
New Delhi
Upadhyay S (2003) Serotonin receptors, agonists and antagonists. Indian J Nucl Med 18:1–11
Villaln C, Centurin D, Valdivia L, De Vries P, Saxena PR (2003) Migraine: pathophysiology,
pharmacology, treatment and future trends. Curr Vasc Pharmacol 1:71–84
Wang H-T, Han F, Shi Y-X (2009) Activity of the 5-HT1A receptor is involved in the alteration of
glucocorticoid receptor in hippocampus and corticotropin-releasing factor in hypothalamus in
SPS rats. Int J Mol Med 24:227–231
Wappler F, Fiege M, Am Esch JS (2001) Pathophysiological role of the serotonin system in
malignant hyperthermia. Br J Anaesth 87:794–798
Wellington K, Jarvis B (2002) Spotlight on rizatriptan in migraine. CNS Drugs 16:715–720
Woolley ML, Marsden CA, Fone KC (2004) 5-ht6 receptors. Curr Drug Targets CNS Neurol
Disord 3:59–79
Wu H, Denna TH, Storkersen JN, Gerriets VA (2019) Beyond a neurotransmitter: the role of
serotonin in inflammation and immunity. Pharmacol Res 140:100–114
Xue W, Fu T, Zheng G, Tu G, Zhang Y, Yang F, Tao L, Yao L, Zhu F (2019) Recent advances and
challenges of the drugs acting on monoamine transporters. Curr Med Chem. https://doi.org/10.
2174/0929867325666181009123218
Ye JH, Ponnudurai R, Schaefer R (2001) Ondansetron: a selective 5-HT3 receptor antagonist and its
applications in CNS-related disorders. CNS Drug Rev 7:199–213
Zarrindast M-R, Nasehi M, Hoseinpourfard M (2014) A mini review of serotonin and its receptors.
Int J Med Rev 1:39–45
Pharmacology of Histamine, Its Receptors
and Antagonists in the Modulation 7
of Physiological Functions
Abstract
Keywords
Histamine · Histamine receptor · Pharmacology · Antagonists · Agonists ·
Biological role · Structural biology · Distribution
B. Gorain (*)
School of Pharmacy, Faculty of Health and Medical Science, Taylor’s University, Subang Jaya,
Selangor, Malaysia
P. Sengupta
Department of Physiology, Faculty of Medicine, Bioscience and Nursing, MAHSA University,
Kuala Lumpur, Malaysia
S. Dutta
Department of Oral Biology and Biomedical Sciences, Faculty of Dentistry, MAHSA University,
Kuala Lumpur, Malaysia
M. Pandey · H. Choudhury
School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
Abbreviations
7.1 Introduction
Histamine, a biologic amine, was isolated from a mould ergot by Sir Henry Dale and
his group at the Wellcome Laboratories more than a century ago. Since then, this
amine was studied enormously to explore associated physiological roles within the
biological system (Parsons and Ganellin 2009). This amine is considered as the most
imperative antique mediator within the biologic system, and it had been studied
extensively among the chemical mediators, including catecholamine and others
derived from amino acids for its role in the biological system (MacGlashan 2003).
Officially histamine was synthesized and characterized as a potential biologic amine
in 1907 and 1910, respectively (Barger and Dale 1910). During initial experiments
with this amine, its capabilities of constricting guinea pig ileum and its convincing
vasopressor potential had been reported. Further studies revealed the stimulatory
7 Pharmacology of Histamine, Its Receptors and Antagonists in the Modulation of. . . 215
To explain the biological role of histamine, it displays two basic and vital
functionalities, viz. imidazole (pKa 9.4) and primary aliphatic amine (pKa 5.8). In
aqueous environment, histamine establishes various acid-base equilibrium. Actually,
the neutral histamine or the free-base is able to receive one or two protons to form the
monocationic or dicationic fraction of histamine, respectively. The formation of
cationic fraction of histamine depends on the pH of the media, where the major
fraction (96%) of histamine forms the monocationic form at physiological pH (7.4),
with 3% dicationic and a small remaining fraction of neutral form. Alternatively, the
predominant form at less than pH 5 is the dicationic, and neutral at pH above
10 (Ramírez et al. 2003; Vianello and Mavri 2012).
Figure 7.1 represents the chemical structure of histamine with nomenclature of
the positions. This nomenclature process of histamine represents its tautomeric
forms, which is important to denote synthesis, storage, release, and metabolism of
histamine, including derivatives of this biological amine.
Histamine is a low molecular weight amine, which is synthesized from the amino
acid L-histidine by the presence of histidine decarboxylase (Huang et al. 2018). Any
other pathway has not been reported to achieve the transformation of histamine
(Parsons and Ganellin 2009). Following purification of histidine decarboxylase
protein from different cells of mouse and rat, a complementary DNA (cDNA) was
cloned that encodes this protein. The gene responsible for histidine decarboxylase
synthesis is Hdc gene, which prepares the proenzyme for the protein. The proenzyme
consists no enzymatic activity encompassing a molecular mass of 74 kDa. Probably
by Caspase-9, the proenzyme is cleaved to form two subunits, 20 kDa C-terminus
and 53 kDa N-terminus subunits. This N-terminal subunit then forms a homodimer
to form the active enzyme, which takes part in catalyse histamine synthesis (Furuta
et al. 2007; Komori et al. 2012). The expression of histidine decarboxylase enzyme
can be observed in several living cells in human body system, including mast cells,
basophils, parietal cells, gastric mucosa, neurons, and in central nervous system
(CNS). Therefore, this enzyme catalyses conversion of histamine from histidine in
different cells and tissues to exert its physiological roles via acting on four different
types of receptors (Oda et al. 2000; Akdis and Blaser 2003; MacGlashan 2003).
Recently, several lymphoid and myeloid cell types have been established with
higher histidine decarboxylase response, which are also capable of synthesizing high
quantities of histamine (Szeberényi et al. 2001). This type of amine is known as
“neo-synthesized histamine”, the presence of which has been located in
haematopoietic progenitors, T cells, dendritic cells, platelets, macrophages, and
neutrophils (Shiraishi et al. 2000; Tanaka et al. 2004; Dy and Schneider 2004;
Shahid et al. 2009).
The preparation of histidine decarboxylase knockout mouse model generates a
system devoid of histamine, where such models are more beneficial to establish the
role of endogenous histamine in a broad range of diseased and normal living system.
Due to lack of histamine, simultaneously a number of mast cells storage granules are
found to decrease in such knockout system, which could be correlated with the
action of histamine on production of mast cells granule proteins (Ohtsu et al. 2001).
It had been shown that in the absence of specific antigens, immunoglobulin E (IgE)
clones activate interleukin-3-dependent BMMC (bone marrow-derived mast cells),
leading towards histamine production, cytokine secretion, adhesion, migration and
improvement of survival time (Kawakami and Kitaura 2005). Simultaneous research
by Tanaka and group added that the transient induction of histidine decarboxylase by
the stimulation of IgE in the BMMC was approximately 200-fold higher than the
production of histidine decarboxylase in the presence of any antigen (Tanaka et al.
2002). Therefore, such induction further increases in the quantity of stored histamine
at the site.
7 Pharmacology of Histamine, Its Receptors and Antagonists in the Modulation of. . . 217
It is evident from the earlier section that the production of histamine may occur in
several tissues within the body. However, binding capabilities to different areas of
the body and subsequent physiological roles are different because of the types of
receptor where it binds to bring the conformational change and physiological
response. Even though the heterogeneity of the histamine receptor had been
established in the 1940s, much information was on histamine-related activities
based on its release, storage and metabolism. Most of the related scientists of British
pharmacology were in agreement, including West GB, Trendelenburg U, Schild HO,
Riley JF, Perry WLM, Paton WDM, Mongar JL, Gaddum JH, Feldberg W and
Blaschko H (Corcoran 1957). Much of the research related to histamine was
compiled together and brought to the researchers worldwide jointly by the British
Pharmacological Society and the Physiological Society in a CIBA Foundation
symposium in honour of Sir Henry Dale (Schayer 1956). The mast cell as a storage
location for histamine was recognized by Riley and West, and it had been established
that certain components are responsible for inducing the release of histamine via
disruption of the mast cells and thereby results in decrease in tissue histamine
content (Riley and West 1952). Therefore, there was a strong positive correlation
established between mast cell population and histamine content in a variety of
tissues. Most of the studies performed initially to release histamine was compound
48/80, a polymer that induces histamine release. It was first described in 1951 by
Paton (Feldberg and Mongar 1954). However, simultaneous comments by Riley and
West made potential changes to the concept that some of the body tissues contained
certain cells other than mast cells to store histamine to account their histamine
content. Thus, subsequent research had brought forward towards those other cells
that store histamine, including blood basophils and platelets in some species
(Parsons and Ganellin 2006). Therefore, mast cells and basophils are the storage
location of histamine in the haematopoietic system, where this histamine could be
found within the specific granules. In mast cells, this amine is closely accompanying
anionic proteoglycans heparin, whereas in basophils it is associated with chondroi-
tin-4-sulphate. The release of a large quantity of histamine is potentiated during
degranulation of the storage granules by the action of immunological response by
IgE or cytokines, or by non-immunological response by calcium ionophore, com-
pound 48/80, substance P, etc. (Dy and Schneider 2004).
The production of histamine within enterochromaffin-like cells also has been
proven and its role in the secretion of gastric juice is established (Dy and Schneider
2004). Therefore, the presence of histamine can be found in all types of tissues
within the biological system of mammalian species; however, the range of concen-
tration of histamine may vary from 1 to over 100 μg/g. Moreover, major storage of
histamine was found in the skin, lung, connective tissues and most of the gastroin-
testinal tract (Parsons and Ganellin 2006).
218 B. Gorain et al.
mepyramine, while other responses were indifferent. Just before two decades, another
two types of histamine receptors, H3 and H4, were identified in consecutive years,
1999 and 2000, respectively (Oda et al. 2000; Nieto-Alamilla et al. 2016). The
structural biology, biological distribution, physiological roles, and available
antagonists are summarized in successive sections of this chapter.
such as in the CNS, gastrointestinal tract, respiratory system and vascular smooth
muscle, hepatocytes, endothelial cells, T and B lymphocytes, dendritic cells,
chondrocytes, monocytes, neutrophils, cardiovascular system, genitourinary system
and adrenal medulla (Chand and Eyre 1975; Hill et al. 1977; Matsuda et al. 2004;
Sander et al. 2006; Shahid et al. 2009). A greater density of H1 receptors has been
shown in the hippocampus, posterior hypothalamus, neocortex, nucleus accumbens
and thalamus. These receptors are found in lower density in the cerebellum and basal
ganglia (Shahid et al. 2009; Mahdy and Webster 2014).
Signalling Pathway
H1 receptors’ established signalling mechanisms include Gq/11 protein activation
along with subsequent activation of phospholipase C (PLC). This leads to increased
intracellular inositol phosphates (IPs) and calcium levels, followed by activation of
small G proteins, RhoA and Rac (Notcovich et al. 2010). Alternatively, in heterolo-
gous native H1 receptors expression systems, the pathway is mediated via Gi/o,
phospholipase A2 activation and production of cyclic guanosine monophosphate
(cGMP). This is followed by nitric oxide production to trigger inflammatory
conditions (Monczor and Fernandez 2016). In certain tissues like the brain and
adrenal glands in mammals, and ovary cells (as seen in Chinese hamster), H1
receptors may also lead to the activation of adenylyl cyclase (AC) followed by
increased intracellular cAMP (30 ,50 -cyclic adenosine monophosphate) production
(Notcovich et al. 2010; Monczor and Fernandez 2016). Besides the signalling
activations by ligand binding, H1 receptors have also been reported to show sponta-
neous receptor activities even when agonists do not bind them. They are able to
activate both IP production and alter gene expression via nuclear factor-κβ
(Fitzsimons et al. 2004; Notcovich et al. 2010; Monczor and Fernandez 2016).
Neurophysiology
Endogenous histamine from neurons that have their cell bodies in the hypothalamic
tuberomammillary nucleus is able to activate the histamine H1 receptors (Haas et al.
2008). These neurons turn active during the “wakefulness” cycle (firing at about
2 Hz), while at the time of slow wave sleep the neurons firing rate reduces to as low
as 0.5 Hz and finally ceases during REM sleep (Thakkar 2011). Thus, the
tuberomammillary nucleus (histaminergic nucleus) plays the major role in regulating
sleep-wakefulness cycle (Sherin et al. 1998). Therefore, these histamine H1
receptors are one of the most important receptors that mediate internal clock. As
histamine acts upon these H1 receptors, it modulates the neurochemistry to trigger
wakefulness and a state of alertness. H1-antihistamines that are able to breach the
blood-brain barrier (BBB) are reported to inhibit H1 receptor activity on the hista-
minergic neurons arising from the tuberomammillary nucleus. This explains the
effect of drowsiness associated with these drugs. In the cerebellum and hippocam-
pus, plentiful histamine H1 receptors have been found in the Purkinje and pyramidal
cells dendrites (Haas et al. 2008). Hippocampal activation of histamine H1 receptors
7 Pharmacology of Histamine, Its Receptors and Antagonists in the Modulation of. . . 221
Inflammation
Stimulation of H1 receptors (other than in the nervous system) attributes to allergic
reactions such as motion sickness, bronchoconstriction, separation of the blood
vessels cell-lining, vasodilatation, redness of skin, hives (skin rashes) and smooth
muscle relaxation. Excessive stimulation of these receptors thus leads to aggravated
allergic progression, such as hay fever and other seasonal allergies. H1 receptor
induced inflammation is mostly mediated via expression of the transcription factor,
NF-κβ. Histamine H1 receptors activation in vascular endothelial cells triggers the
production and release of many neuromodulators, such as platelet-activating factor,
prostacyclin and nitric oxide (Sharma 2004). H1 receptor activation can also cause
vascular permeability alterations, specifically in the postcapillary venule by contrac-
tion of endothelial cells (Sharma 2004; Thurmond et al. 2008; Criado et al. 2010).
H1-antihistamines have been reported to downregulate NF-κβ expression, thereby
attenuating certain inflammatory processes (Church and Church 2011; Church
2017).
First-Generation H1-Antihistamines
The first-generation H1-antihistamines share similar chemical constituents with
cholinergic muscarinic antagonists, antipsychotics, tranquilizers and antihyperten-
sive drugs (Mahdy and Webster 2014). These similarities render them poorly
selective leading to undesired cross-talks with other receptors and often causing
anti-α-adrenergic, antimuscarinic, as well as antiserotonin effects. They are potent to
cross the BBB and thereby can adversely affect histaminergic transmission (Church
and Church 2011).
Physiologically, histamine release during the daytime leads to wakefulness or
state of alert while the reduced levels at night account for passive decrease in arousal
response. The first-generation H1-antihistamines during the day often result in
daytime drowsiness and impaired concentration. At night, these drugs delay the
onset of REM sleep and decrease its duration (Rojas-Zamorano et al. 2009). The lack
222 B. Gorain et al.
Fig. 7.2 Conformational changes in histamine H1 receptor on binding with agonists and
antagonists
Second-Generation H1-Antihistamines
The 1980s had witnessed a major advance in antihistaminergic drug development
with the introduction of the second-generation H1-antihistamines (Criado et al.
2010). These possess limited access across the BBB rendering them less sedating
as compared to the first-generation H1-antihistamine. Moreover, they are more
ligand specific and have no anticholinergic effects (Church and Church 2011).
Usually, two factors determine the net efficacy of H1-antihistaminer agents, one
being its receptor affinity or absolute potency and the other is the drug concentration
at the receptors site. It has been reported that desloratadine is the most effective
antihistamine (Ki 0.4 nM), followed by levocetirizine (Ki 3 nM) and fexofenadine
7 Pharmacology of Histamine, Its Receptors and Antagonists in the Modulation of. . . 223
(Ki 10 nM). These drugs attain higher potency in lower concentration. Physical
conditions like temperature and pH may modulate the efficacy of these drugs in
physiologic and pathologic conditions. For example, the H1 receptor affinity of
fexofenadine and levocetirizine is increased even by fivefold in inflammatory
conditions when tissue pH drops (Church and Church 2011; Mahdy and Webster
2014).
Several factors influence the free drug concentrations in the tissue compartments
and extracellular fluids. In this regard, their absorption into the systemic circulation
followed by ingestion of tablet or capsule or other oral dosage form containing the
drug is vital. H1-antihistamines are properly absorbed, with the exception of
fexofenadine which has variable absorption due to the effects of active transporter
proteins (Devillier et al. 2008). Another important factor is the plasma binding of the
drugs, which in case of this group is quite high (~65% with desloratadine, ~90%
with levocetirizine) (Church and Church 2011). Finally, the volume of distribution
of the drugs throughout the body influences its plasma levels after the distribution.
For example, levocetirizine (0.4 L/kg) has low body distribution potency, while
fexofenadine (5.4–5.8 L/kg) and desloratadine (~49 L/kg) are highly distributed
throughout the body tissues (Molimard et al. 2004). Since H1-antihistamines
concentrations in specific extracellular fluids are difficult to obtain, an indirect
estimate of their efficacy can be calculated using the data on its receptor occupancy
(absolute potency) and its peak plasma concentrations generally at ~4 h followed by
one oral dose:
Receptor occupancy (%) ¼ Bmax L + Ki, where Bmax: maximal number of
binding sites (set to 100%); L: plasma free drug concentration; Ki: equilibrium
inhibition constant or absolute potency (Gillard et al. 2005).
The time required for onset of drug action correlates with its oral absorption rate,
but this straight relation on speed of onset of antihistamine actions with its oral
absorption rate is often breached by several confounding factors. In a study, it has
been shown that plasma concentration of levocetirizine reached after 30 min from its
injection in children with histamine-induced flare response. However, the drug had
taken more than one and half-hour for its diffusion into the extravascular space for
utmost clinical effect. Most of the H1-antihistamines take around 4 h to inhibit such
flare responses to the maximal limits in adults (Tashiro et al. 2009).
Certain anti-H1-histamines, like levocetirizine and desloratadine, possess longer
systemic action in mitigating histamine-induced flare responses than their predicted
duration of action from their plasma concentrations (Tashiro et al. 2009). This
presumably occurs owing to drug “trapping” by the H1 receptors by strong and
enduring binding (Church and Church 2011). Alternatively, fexofenadine
possesses comparatively shorter duration action. The primary reason behind this less
prolonged action of fexofenadine is that it is actively secreted into the intestine and
urine by P-glycoprotein (Tannergren et al. 2003).
A moderate quantity of research on H1-antihistamines has been centred on the
early phase allergic responses of histamines, but recent studies focus upon the anti-
inflammatory effects of these drugs (Boyle et al. 2006; Church and Church 2011).
H1-antihistamines have been seen to attenuate inflammations owing to nasal
224 B. Gorain et al.
Ash and Schild proposed the existence of two classes of histamine receptors, H1 and
H2, upon introduction of selective ligands for α- and β-adrenergic receptors and
subsequently Black proved the concept following research outcomes with selective
ligands for β-adrenergic receptors and later accepted upon synthesis of selective H2
blockers. The histamine H2 receptor is a Gs-coupled GPCRs and was firstly defined
pharmacologically by Sir James (Church 2009; Timmerman and van der Goot
2009).
Histamine H3 receptor expressions are mostly predominant in the CNS rather than in
the peripheral nervous system. These receptors function as autoreceptors in presyn-
aptic histaminergic neurons (Nieto-Alamilla et al. 2016). H3 receptors also mediate
histamine turnover via negative feedback to inhibit the synthesis and release of
histamine. These also lead to feedback inhibition of several other neurotransmitters
released by functioning as an inhibitory heteroreceptor. These neurotransmitters
include gamma amino-butyric acid (GABA), dopamine, noradrenaline, acetylcho-
line, serotonin and histamine. The H3 receptors gene sequences possess as less as
about 22% and 20% homology with H1 and H2 histamine receptors, respectively.
The H3 receptor is a potential therapeutic target due to its regulatory functions in
several neuronal mechanisms (Nieto-Alamilla et al. 2016).
Fig. 7.4 Molecular signalling pathway of activated histamine H3 receptor (H3R) located in
presynaptic and postsynaptic neuron to mediate biological functions. AC adenylyl cyclase; MAPK
mitogen-activated protein kinases; PI3K phosphoinositide 3-kinase; PLC phospholipase C; PLA2
phospholipase A2; GIRK G-protein-coupled inwardly rectifying potassium channel
Activation of Phospholipase C
Histamine H3 receptors may activate phospholipase C (PLC), thereby leading to
increased intracellular calcium ions concentration via IP3 pathway to mediate
physiological functions (Bongers et al. 2007).
230 B. Gorain et al.
Stimulation of Phospholipase A2
Histamine H3 receptor-induced phospholipase A2 activation leads to the release of
docosahexaenoic acid, arachidonic acid and lysophospholipids. These mediate
physiological functions such as relaxation of bronchioles via endothelium-derived
relaxing factor, which is an arachidonic acid metabolite (Nieto-Alamilla et al. 2016).
Early Pharmacophore
The initial focus of H3 receptor ligands development was upon the agonists with an
imidazole ring within its structure (Celanire et al. 2005). The imidazole ring in these
drugs leads to undesired inhibition of cytochrome P450 isoenzymes and this results
in adverse drug interactions (Celanire et al. 2005). Moreover, they fail to cross the
BBB and also proved to have a certain degree of toxicity (Schwartz 2011). Another
problem with these drugs is less specificity and action upon other receptors such as
on the histamine H4 receptor (Sadek et al. 2016).
Thioperamide represents the first imidazole-based histamine H3 antagonist. It
was quite potent as well as selective in action but could cause hepatotoxicity. It was
first designed in order to enhance cognition functions and wakefulness (Schwartz
2011). A potential study has reported the efficacy of thioperamide treatment in
ameliorating circadian rhythm Parkinson patient (Masini et al. 2017).
New Pharmacophore
To mitigate the limitations of histamine H3 receptor imidazole based antagonists,
non-imidazole H3 receptor antagonists emerged. These drugs can easily cross the
BBB and reach the CNS. However, few problems also cropped up in the use of these
drugs, for example phospholipidosis, its strong binding to potassium channel, and
problems with P-glycoprotein also known as multidrug resistance protein 1 (MDR1)
substrate (Gemkow et al. 2009). Pitolisant, a highly selective antagonist for the H3
receptor, was the first H3 receptor antagonist to proceed to clinical trials. Presently, it
is the sole drug that is approved in the USA and Europe.
7 Pharmacology of Histamine, Its Receptors and Antagonists in the Modulation of. . . 231
The gene of H4 receptor was discovered in 1999 from the Human Genome Project,
which had a feature of class A GPCR. This receptor is found to have similarity with
other histamine receptors in its homology; thus it became the fourth receptor in the
histamine family. After the huge success of H1 and H2 targeting, H4 was also
underwent investigation for its targetability and functions. A vast literature review
revealed that some of the histaminic activities were not mediated by H1, H2 and H3
receptors. In this context, pruritus and asthma came up with the major area of
investigation as histamine plays an important role in the pathogenesis of disease.
However, H1 and H2 targeted histamines does not respond effectivity for these
disease conditions. Later this gap was filled with the concept of H4 receptor media-
tion in inflammation and lung functions. Likewise, patients with atopic dermatitis
were not responding to H1 receptor antagonists. Subsequently, the generation of H4
antagonist has shown tremendous effect on disease conditions (Leurs et al. 2012;
Thurmond 2015; Kiss and Keserű 2016).
binding but play significant roles in receptor activation (Shin et al. 2002). Another
study for homology model development confirmed Asp943.3.2 and Glu1825.4.6 as the
major anchoring point for H4 receptor binding and has shown participation in GPCR
ligand binding. Moreover, Asp943.3.2 is the major site for ligand interaction and
situated closer to Glu1825.4.6 (Evers and Klabunde 2005; Kiss et al. 2008a; Pontiki
and Hadjipavlou-Litina 2017). The binding mode of antagonists and agonists was
also investigated by using histamine as agonist and JNJ7777120 as antagonist.
Experimental results revealed that antagonists interact with Asp943.3.2 and
Glu1825.4.6, whereas agonists interact with Thr3236.5.5, Asp943.3.2 and Glu1825.4.6
which suggest the involvement of Thr3236.5.5 in agonist binding and activation of
receptors (Jablonowski et al. 2003; Kiss et al. 2008b; Jójárt et al. 2008).
Fig. 7.5 Indicative immunomodulatory actions of histamine that are mediated through histamine
H4 receptors (H4) predominately expressed in immune cells. Gαi/o, G-protein; TH, helper T cell
(Zampeli and Tiligada 2009)
downregulate the production of ILs and that of CCL2 (C-C motif chemokine ligand
2) in human monocyte-derived inflammatory dendritic epidermal cells, the later
leading to decreased monocyte migration (Dijkstra et al. 2007, 2008).
Diseases like asthma and atopic dermatitis are associated with mast cells and
eosinophils. Histamine H4 receptors act as a mediator in lung disorders by
controlling the migration of inflammatory cells, production of chemokine and
cytokine. The expression of histamine H4 receptor in the lung is low; however,
high expression in smooth cells and bronchial epithelial contributes to disease
phenotype (Gantner et al. 2002). As discussed earlier, it also mediated the recruit-
ment and distribution of mast cells in bronchial epithelium, thus leading to allergic
inflammation of the airways (Thurmond et al. 2004). This was evident by a study on
guinea pig model, where the H4 receptor antagonist JNJ7777120 showed reduction
in inflammation, improved lung function and inflammation mediators synthesis in
the lungs after allergen challenge (Somma et al. 2013). Histamine H4 receptor
involvement is also recognized in autoimmune disease, such as rheumatoid arthritis
(RA). Further, RA severity could be related to the expression of H4 receptor along
with H1 and H2. This concept was supported by the presence of H4 receptor in
vascular wall cells and synovial tissue of the patients suffering with RA and
osteoarthritis (Ohki et al. 2007; Kiss et al. 2008a).
234 B. Gorain et al.
7.4 Conclusion
References
Akdis CA, Blaser K (2003) Histamine in the immune regulation of allergic inflammation. J Allergy
Clin Immunol 112:15–22
Anon (2009) Joint formulary committee. In: British national formulary, 57rd edn. British Medical
Association and Royal Pharmaceutical Society of Great Britain, London
7 Pharmacology of Histamine, Its Receptors and Antagonists in the Modulation of. . . 235
Arrang J-M, Garbarg M, Schwartz J-C (1983) Auto-inhibition of brain histamine release mediated
by a novel class (H3) of histamine receptor. Nature 302:832–837
Arrang J-M, Garbarg M, Lancelo J-C et al (1987) Highly potent and selective ligands for histamine
H3-receptors. Nature 327:117–123
Arrang J-M, Morisset S, Gbahou F (2007) Constitutive activity of the histamine H3 receptor.
Trends Pharmacol Sci 28:350–357
Ash AS, Schild HO (1966) Receptors mediating some actions of histamine. Br J Pharmacol
Chemother 27:427–439
Atzori M, Lau D, Tansey EP et al (2000) H2 histamine receptor-phosphorylation of Kv3.2
modulates interneuron fast spiking. Nat Neurosci 3:791–798
Barger G, Dale H (1910) The presence of ergot and physiological activity of Beta-
imidazolylethylamine. J Physiol 40:xxxviii–xl
Barnard R, Barnard A, Salmon G et al (2008) Histamine-induced actin polymerization in human
eosinophils: an imaging approach for histamine H 4 receptor. Cytom Part A 73A:299–304
Barnes PJ, Cuss FM, Palmer JB (1985) The effect of airway epithelium on smooth muscle
contractility in bovine trachea. Br J Pharmacol 86:685–691
Benarroch EE (2010) Histamine in the CNS: multiple functions and potential neurologic
implications. Neurology 75:1472–1479
Best CH, Dale HH, Dudley HW, Thorpe WV (1927) The nature of the vaso-dilator constituents of
certain tissue extracts. J Physiol 62:397–417
Birdsall NJ (1991) Cloning and structure-function of the H2 histamine receptor. Trends Pharmacol
Sci 12:9–10
Black JW, Duncan WA, Durant CJ et al (1972) Definition and antagonism of histamine H
2 -receptors. Nature 236:385–390
Bongers G, Bakker RA, Leurs R (2007) Molecular aspects of the histamine H3 receptor. Biochem
Pharmacol 73:1195–1204
Boyle J, Eriksson M, Stanley N et al (2006) Allergy medication in Japanese volunteers: treatment
effect of single doses on nocturnal sleep architecture and next day residual effects. Curr Med Res
Opin 22:1343–1351
Brimblecombe RW, Duncan WA, Durant GJ et al (1975) The pharmacology of cimetidine, a new
histamine H2-receptor antagonist. Br J Pharmacol 53:435P–436P
Brioni JD, Esbenshade TA, Garrison TR et al (2011) Discovery of histamine H3 antagonists for the
treatment of cognitive disorders and Alzheimer’s disease. J Pharmacol Exp Ther 336:38–46
Celanire S, Wijtmans M, Talaga P et al (2005) Keynote review: histamine H3 receptor antagonists
reach out for the clinic. Drug Discov Today 10:1613–1627
Chand N, Eyre P (1975) Classification and biological distribution of histamine receptor sub-types.
Agents Actions 5:277–295
Church MK (2009) Histamine and its receptors. In: Allergy Frontiers: classification and
Pathomechanisms. Springer, Tokyo, pp 329–356
Church MK (2017) Allergy, histamine and antihistamines. In: Handbook of experimental pharma-
cology. Springer, New York, NY, pp 321–331
Church DS, Church MK (2011) Pharmacology of antihistamines. World Allergy Organ J 4:S22–
S27
Corcoran AC (1957) Histamine. Ciba foundation symposium jointly with the Physiological Society
and the British pharmacological Society in Honor of sir Henry Dale. Arch Intern Med
99:157–158
Cowden JM, Zhang M, Dunford PJ, Thurmond RL (2010) The histamine H4 receptor mediates
inflammation and pruritus in Th2-dependent dermal inflammation. J Invest Dermatol
130:1023–1033
Criado PR, Criado RFJ, Maruta CW, Machado FC (2010) Histamine, histamine receptors and
antihistamines: new concepts. An Bras Dermatol 85:195–210
Dale HH (1929) Some chemical factors in the control of the circulation. Lancet 1:167–223
236 B. Gorain et al.
Hill SJ (1990) Distribution, properties, and functional characteristics of three classes of histamine
receptor. Pharmacol Rev 42:45–83
Hill SJ, Young JM, Marrian DH (1977) Specific binding of 3H-mepyramine to histamine H1
receptors in intestinal smooth muscle. Nature 270:361–363
Hill SJ, Ganellin CR, Timmerman H et al (1997) International Union of Pharmacology. XIII.
Classification of histamine receptors. Pharmacol Rev 49:253–278
Hofstra CL, Desai PJ, Thurmond RL, Fung-Leung W-P (2003) Histamine H4 receptor mediates
chemotaxis and calcium mobilization of mast cells. J Pharmacol Exp Ther 305:1212–1221
Hsieh GC, Chandran P, Salyers AK et al (2010) H4 receptor antagonism exhibits anti-nociceptive
effects in inflammatory and neuropathic pain models in rats. Pharmacol Biochem Behav
95:41–50
Huang Z-L, Mochizuki T, Qu W-M et al (2006) Altered sleep-wake characteristics and lack of
arousal response to H3 receptor antagonist in histamine H1 receptor knockout mice. Proc Natl
Acad Sci 103:4687–4692
Huang H, Li Y, Liang J, Finkelman FD (2018) Molecular regulation of histamine synthesis. Front
Immunol 9:1392
Ishizaka T, De Bernardo R, Tomioka H et al (1972) Identification of basophil granulocytes as a site
of allergic histamine release. J Immunol 108:1000–1008
Jablonowski JA, Grice CA, Chai W et al (2003) The first potent and selective non-imidazole human
histamine H4 receptor antagonists. J Med Chem 46:3957–3960
Jójárt B, Kiss R, Viskolcz B, Keseru GM (2008) Activation mechanism of the human histamine H4
receptor--an explicit membrane molecular dynamics simulation study. J Chem Inf Model
48:1199–1210
Jemima EA, Prema A, Thangam EB (2014) Functional characterization of histamine H4 receptor on
human mast cells. Mol Immunol 62:19–28. https://doi.org/10.1016/j.molimm.2014.05.007
Kawakami T, Kitaura J (2005) Mast cell survival and activation by IgE in the absence of antigen: a
consideration of the biologic mechanisms and relevance. J Immunol 175:4167–4173
Kiss R, Keserű GM (2016) Structure-based discovery and binding site analysis of histamine
receptor ligands. Expert Opin Drug Discov 11:1165–1185
Kiss R, Kiss B, Könczöl A et al (2008a) Discovery of novel human histamine H4 receptor ligands
by large-scale structure-based virtual screening. J Med Chem 51:3145–3153
Kiss R, Noszál B, Rácz A et al (2008b) Binding mode analysis and enrichment studies on homology
models of the human histamine H4 receptor. Eur J Med Chem 43:1059–1070
Knight DA, Adcock JA, Phillips MJ, Thompson PJ (1990) The effect of epithelium removal on
human bronchial smooth muscle responsiveness to acetylcholine and histamine. Pulm
Pharmacol 3:198–202
Komori H, Nitta Y, Ueno H, Higuchi Y (2012) Structural study reveals that Ser-354 determines
substrate specificity on human histidine decarboxylase. J Biol Chem 287:29175–29183
Kostopoulos G, Psarropoulou C, Haas HL (1988) Membrane properties, response to amines and to
tetanic stimulation of hippocampal neurons in the genetically epileptic mutant mouse tottering.
Exp Brain Res 72:45–50
Lai X, Ye L, Liao Y et al (2016) Agonist-induced activation of histamine H3 receptor signals to
extracellular signal-regulated kinases 1 and 2 through PKC-, PLD-, and EGFR-dependent
mechanisms. J Neurochem 137:200–215
Leurs R, Bakker RA, Timmerman H, de Esch IJP (2005) The histamine H3 receptor: from gene
cloning to H3 receptor drugs. Nat Rev Drug Discov 4:107–120
Leurs R, Hough LB, Blandina P, Haas HL (2012) Histamine. In: Basic neurochemistry, 8rd edn.
Elsevier, pp 323–341
Lewis T, Grant RT (1924) Vascular reactions of the skin to injury. Part 11. The liberation of
histamine-like substance in the injured skin, the underlying cause of factitious urticaria and of
wheals produced by burning: and observations upon the nervous control of certain skin
reactions. Heart 11:209–265
238 B. Gorain et al.
Lin J-S, Sergeeva OA, Haas HL (2011) Histamine H3 receptors and sleep-wake regulation. J
Pharmacol Exp Ther 336:17–23
Lippert U, Artuc M, Grützkau A et al (2004) Human skin mast cells express H2 and H4, but not H3
receptors. J Invest Dermatol 123:116–123
Lovenberg TW, Roland BL, Wilson SJ et al (1999) Cloning and functional expression of the human
histamine H3 receptor. Mol Pharmacol 55:1101–1107
MacGlashan D (2003) Histamine: a mediator of inflammation. J Allergy Clin Immunol 112:S53–
S59
Mahdy AM, Webster NR (2014) Histamine and antihistamines. Anaesth Intensive Care Med
15:250–255
Masini D, Lopes-Aguiar C, Bonito-Oliva A et al (2017) The histamine H3 receptor antagonist
thioperamide rescues circadian rhythm and memory function in experimental parkinsonism.
Transl Psychiatry 7:e1088–e1088
Matsuda N, Jesmin S, Takahashi Y et al (2004) Histamine H1 and H2 receptor gene and Protein
levels are differentially expressed in the hearts of rodents and humans. J Pharmacol Exp Ther
309:786–795
Meneses D, Mateos V, Islas G, Barral J (2015) G-protein-coupled inward rectifier potassium
channels involved in corticostriatal presynaptic modulation. Synapse 69:446–452
Molimard M, Diquet B, Benedetti MS (2004) Comparison of pharmacokinetics and metabolism of
desloratadine, fexofenadine, levocetirizine and mizolastine in humans. Fundam Clin Pharmacol
18:399–411
Monczor F, Fernandez N (2016) Current knowledge and perspectives on histamine H1 and H2
receptor pharmacology: functional selectivity, receptor crosstalk, and repositioning of classic
histaminergic ligands. Mol Pharmacol 90:640–648
Morisset S, Rouleau A, Ligneau X et al (2000) High constitutive activity of native H3 receptors
regulates histamine neurons in brain. Nature 408:860–864
Nakayama T, Kato Y, Hieshima K et al (2004) Liver-expressed chemokine/CC chemokine ligand
16 attracts eosinophils by interacting with histamine H4 receptor. J Immunol 173:2078–2083
Nakazawa H, Sekizawa K, Morikawa M et al (1994) Viral respiratory infection causes airway
hyperresponsiveness and decreases histamine N-methyltransferase activity in Guinea pigs. Am J
Respir Crit Care Med 149:1180–1185
Nieto-Alamilla G, Marquez-Gomez R, Garcia-Galvez A-M et al (2016) The histamine H3 receptor:
structure, pharmacology, and function. Mol Pharmacol 90:649–673
Notcovich C, Diez F, Tubio MR et al (2010) Histamine acting on H1 receptor promotes inhibition
of proliferation via PLC, RAC, and JNK-dependent pathways. Exp Cell Res 316:401–411
Oda T, Morikawa N, Saito Y et al (2000) Molecular cloning and characterization of a novel type of
histamine receptor preferentially expressed in leukocytes. J Biol Chem 275:36781–36786
Ogasawara M, Yamauchi K, Satoh Y-I et al (2006) Recent advances in molecular pharmacology of
the histamine systems: organic cation transporters as a histamine transporter and histamine
metabolism. J Pharmacol Sci 101:24–30
Ohki E, Suzuki M, Aoe T et al (2007) Expression of histamine H4 receptor in synovial cells from
rheumatoid arthritic patients. Biol Pharm Bull 30:2217–2220
Ohtsu H, Tanaka S, Terui T et al (2001) Mice lacking histidine decarboxylase exhibit abnormal
mast cells. FEBS Lett 502:53–56
Panula P, Chazot PL, Cowart M et al (2015) International Union of Basic and Clinical Pharmacol-
ogy. XCVIII. Histamine receptors. Pharmacol Rev 67:601–655
Parks GS, Olivas ND, Ikrar T et al (2014) Histamine inhibits the melanin-concentrating hormone
system: implications for sleep and arousal. J Physiol 592:2183–2196
Parsons ME, Ganellin CR (2006) Histamine and its receptors. Br J Pharmacol 147:S127–S135
Parsons ME, Ganellin CR (2009) Histamine and its receptors. Br J Pharmacol 147:S127–S135
Passani MB, Blandina P, Torrealba F (2011) The histamine H3 receptor and eating behavior. J
Pharmacol Exp Ther 336:24–29
7 Pharmacology of Histamine, Its Receptors and Antagonists in the Modulation of. . . 239
Pedarzani P, Storm JF (1995) Protein kinase A-independent modulation of ion channels in the brain
by cyclic AMP. Proc Natl Acad Sci U S A 92:11716–11720
Pontiki E, Hadjipavlou-Litina D (2017) QSAR models on H 4 receptor antagonists associated with
inflammation and anaphylaxis. J Biomol Struct Dyn 35:968–1005
Raible DG, Lenahan T, Fayvilevich Y et al (1994) Pharmacologic characterization of a novel
histamine receptor on human eosinophils. Am J Respir Crit Care Med 149:1506–1511
Ramírez FJ, Tunon I, Collado JA, Silla E (2003) Structural and vibrational study of the Tautomer-
ism of histamine Free-Base in solution. J Am Chem Soc 125:2328–2340
Reher TM, Neumann D, Buschauer A, Seifert R (2012) Incomplete activation of human eosinophils
via the histamine H4-receptor: evidence for ligand-specific receptor conformations. Biochem
Pharmacol 84:192–203
Riley JF (1953) Histamine in tissue mast cells. Science (80-) 118:332–333
Riley JF, West GB (1952) Histamine in tissue mast cells. J Physiol 117:72P–73P
Rojas-Zamorano JA, Esqueda-Leon E, Jimenez-Anguiano A et al (2009) The H1 histamine receptor
blocker, chlorpheniramine, completely prevents the increase in REM sleep induced by immobi-
lization stress in rats. Pharmacol Biochem Behav 91:291–294
Rouleau A, Ligneau X, Tardivel-Lacombe J et al (2002) Histamine H3-receptor-mediated [35S]
GTPγ[S] binding: evidence for constitutive activity of the recombinant and native rat and human
H3 receptors. Br J Pharmacol 135:383–392
Ruat M, Bouthenet ML, Schwartz JC, Ganellin CR (1990) Histamine H1-receptor in heart: unique
electrophoretic mobility and autoradiographic localization. J Neurochem 55:379–385
Sadek B, Saad A, Sadeq A et al (2016) Histamine H3 receptor as a potential target for cognitive
symptoms in neuropsychiatric diseases. Behav Brain Res 312:415–430
Sahlholm K, Nilsson J, Marcellino D et al (2012) Voltage sensitivities and deactivation kinetics of
histamine H3 and H4 receptors. Biochim Biophys Acta Biomembr 1818:3081–3089
Sander LE, Lorentz A, Sellge G et al (2006) Selective expression of histamine receptors H1R, H2R,
and H4R, but not H3R, in the human intestinal tract. Gut 55:498–504
Schayer RW (1956) The origin and fate of histamine in the body. In: Wolstenholme GEW,
O’Connor CM (eds) Ciba foundation symposium on histamine. J and A Churchill Ltd.,
London, pp 183–188
Schwartz J-C (2011) The histamine H3 receptor: from discovery to clinical trials with pitolisant. Br
J Pharmacol 163:713–721
Seifert R, Schneider EH, Dove S et al (2011) Paradoxical stimulatory effects of the “standard”
histamine H4-receptor antagonist JNJ7777120: the H4 receptor joins the Club of 7 transmem-
brane domain receptors exhibiting functional selectivity. Mol Pharmacol 79:631–638
Sekizawa K, Nakazawa H, Ohrui T et al (1993) Histamine N-methyltransferase modulates hista-
mine- and antigen-induced bronchoconstriction in Guinea pigs In Vivo. Am Rev Respir Dis
147:92–96
Shahid M, Tripathi T, Sobia F et al (2009) Histamine, histamine receptors, and their role in
immunomodulation: an updated systematic review. Open Immunol J 2:9–41
Sharma HS (2004) Histamine influences the blood-spinal cord and brain barriers following injuries
to the central nervous system. Blood Spinal Cord Brain Barriers Health Dis:159–189. https://
doi.org/10.1016/B978-012639011-7/50017-6
Sherin JE, Elmquist JK, Torrealba F, Saper CB (1998) Innervation of histaminergic
tuberomammillary neurons by GABAergic and galaninergic neurons in the ventrolateral
preoptic nucleus of the rat. J Neurosci 18:4705–4721
Shimamura T, Shiroishi M, Weyand S et al (2011) Structure of the human histamine H1 receptor
complex with doxepin. Nature 475:65–70
Shin N, Coates E, Murgolo NJ et al (2002) Molecular modeling and site-specific mutagenesis of the
histamine-binding site of the histamine H4 receptor. Mol Pharmacol 62:38–47
Shiraishi M, Hirasawa N, Oikawa S et al (2000) Analysis of histamine-producing cells at the late
phase of allergic inflammation in rats. Immunology 99:600–606
240 B. Gorain et al.
Simons KJ, Benedetti MS, Simons FER et al (2007) Relevance of H1-receptor occupancy to
H1-antihistamine dosing in children. J Allergy Clin Immunol 119:1551–1554
Somma T, Cinci L, Formicola G et al (2013) A selective antagonist of histamine H 4 receptors
prevents antigen-induced airway inflammation and bronchoconstriction in Guinea pigs: involve-
ment of lipocortin-1. Br J Pharmacol 170:200–213
Sundvik M, Kudo H, Toivonen P et al (2011) The histaminergic system regulates wakefulness and
orexin/hypocretin neuron development via histamine receptor H1 in zebrafish. FASEB J
25:4338–4347
Szeberényi JB, Pállinger E, Zsinkó M et al (2001) Inhibition of effects of endogenously synthesized
histamine disturbs in vitro human dendritic cell differentiation. Immunol Lett 76:175–182
Takeshita K, Sakai K, Bacon KB, Gantner F (2003) Critical role of histamine H4 receptor in
leukotriene B4 production and mast cell-dependent neutrophil recruitment induced by Zymosan
in vivo. J Pharmacol Exp Ther 307:1072–1078
Tanaka S, Takasu Y, Mikura S et al (2002) Antigen-independent induction of histamine synthesis
by immunoglobulin E in mouse bone marrow–derived mast cells. J Exp Med 196:229–235
Tanaka S, Deai K, Konomi A et al (2004) Expression of L-histidine decarboxylase in granules of
elicited mouse polymorphonuclear leukocytes. Eur J Immunol 34:1472–1482
Tannergren C, Petri N, Knutson L et al (2003) Multiple transport mechanisms involved in the
intestinal absorption and first-pass extraction of fexofenadine. Clin Pharmacol Ther 74:423–436
Tashiro M, Kato M, Miyake M et al (2009) Dose dependency of brain histamine H1 receptor
occupancy following oral administration of cetirizine hydrochloride measured using PET with
[11 C]doxepin. Hum Psychopharmacol Clin Exp 24:540–548
Thakkar MM (2011) Histamine in the regulation of wakefulness. Sleep Med Rev 15:65–74
Thurmond RL (2015) The histamine H4 receptor: from orphan to the clinic. Front Pharmacol 6:65
Thurmond RL, Desai PJ, Dunford PJ et al (2004) A potent and selective histamine H4 receptor
antagonist with anti-inflammatory properties. J Pharmacol Exp Ther 309:404–413
Thurmond RL, Gelfand EW, Dunford PJ (2008) The role of histamine H1 and H4 receptors in
allergic inflammation: the search for new antihistamines. Nat Rev Drug Discov 7:41–53
Thurmond R, Wolin RL, Wei J et al (2009) Pharmacological characterization of oxime agonists of
the histamine H4 receptor. J Receptor Ligand Channel Res 3:37
Thurmond RL, Greenspan A, Radziszewski W et al (2016) Toreforant, a histamine H4 receptor
antagonist, in patients with active rheumatoid arthritis despite methotrexate therapy: results of
2 phase II studies. J Rheumatol 43:1637–1642
Timmerman H, van der Goot H (2009) Histamine receptors and their ligands: mechanisms and
applications. In: Encyclopedia neuroscience. Springer, New York, NY, pp 1149–1166
Traiffort E, Ruat M, Arrang JM et al (1992) Expression of a cloned rat histamine H2 receptor
mediating inhibition of arachidonate release and activation of cAMP accumulation. Proc Natl
Acad Sci 89:2649–2653
Traiffort E, Vizuete ML, Tardivellacombe J et al (1995) The Guinea pig histamine H2 receptor:
gene cloning, tissue expression and chromosomal localization of its human counterpart.
Biochem Biophys Res Commun 211:570–577
Varga C, Horvath K, Berko A et al (2005) Inhibitory effects of histamine H4 receptor antagonists on
experimental colitis in the rat. Eur J Pharmacol 522:130–138
Vianello R, Mavri J (2012) Microsolvation of the histamine monocation in aqueous solution: the
effect on structure, hydrogen bonding ability and vibrational spectrum. New J Chem 36:954
Yu F, Wolin RL, Wei J et al (2010) Pharmacological characterization of oxime agonists of the
histamine H4 receptor. J Receptor Ligand Channel Res 3:37–49. https://doi.org/10.2147/
JRLCR.S6468
Zampeli E, Tiligada E (2009) The role of histamine H4 receptor in immune and inflammatory
disorders. Br J Pharmacol 157:24–33
Zamponi GW, Currie KPM (2013) Regulation of ca(V)2 calcium channels by G protein coupled
receptors. Biochim Biophys Acta 1828:1629–1643
Pharmacology of GABA and Its Receptors
8
Sunpreet Kaur, Shamsher Singh, Anchal Arora, Parladh Ram,
Sachin Kumar, Puneet Kumar, and Sara Nidal Abed
Abstract
S. Kaur · S. Singh
Neuroscience Division, Department of Pharmacology, ISF College of Pharmacy, Moga,
Punjab, India
A. Arora · P. Ram · S. Kumar
Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical
University, Bathinda, Punjab, India
P. Kumar (*)
Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical
University, Bathinda, Punjab, India
S. N. Abed
Faculty of Pharmacy, Philadelphia University, Amman, Jordan
sclerosis (MS), and schizophrenia. This chapter further highlights the recent and
previously conducted clinical trials and future investigational targets of GABA.
Now it is readily accepted as a vital spinal and supra-spinal inhibitory transmitter
and we know many details regarding its molecular structure and trafficking
around neurons. The chapter highlights the synthesis, reuptake, and metabolism
of GABA. Thereafter, mechanisms of action of various GABA subtypes
(GABAA, GABAB, GABAC) have been discussed which make the prospects
for further research very exciting.
Keywords
Basal ganglia · GABA · Hyperpolarization · Neurological disorders · Inhibition
Abbreviations
AC Adenylyl cyclase
AD Alzheimer’s disease
AEDs Antiepileptic drugs
BGT Betaine-GABA transporter
BZD Benzodiazepine
cAMP Cyclic adenosine monophosphate
CB Cannabinoid receptor
CNS Central nervous system
D Dopamine
DSM Diagnostic and Statistical Manual of Mental Disorder
FSH Follicle stimulating hormone
GABA Gamma aminobutyric acid
GAD Glutamic acid decarboxylase
GAD Generalized anxiety disorder
GATs GABA transporters
GDNF Glial cell derived neurotrophic factor
GHB Gamma-hydroxybutyrate
Gln Glutamine
GnRH Gonadotropin releasing hormone
GPCR G-protein-coupled receptor
Gpe Globus pallidus external
Gpi Globus pallidus internal
HD Huntington’s disease
IN Interneuron
IPSPs Inhibitory postsynaptic potentials
mBDNF mature Brain-derived neurotrophic factor
MnPO Median preoptic nuclei
MNTB Medial nucleus of the trapezoid body
MPN Methylpyridoxine
8 Pharmacology of GABA and Its Receptors 243
MS Multiple sclerosis
NMDA N-methyl-D-aspartate
nNOS Neuronal nitric oxide synthase
OCD Obsessive compulsive disorder
PAG Phosphate-activated glutaminase
PD Panic disorder
PD Parkinson’s disease
PFC Prefrontal cortex
PLTS Persistent low-threshold spiking
PPMS Primary progressive multiple sclerosis
PRMS Progressive relapsing multiple sclerosis
PTSD Post-traumatic stress disorder
RIMS Rapid eye movement sleep
RRMS Relapsing remitting multiple sclerosis
SACs Starburst amacrine cells
SAD Social anxiety disorder
SCZ Schizophrenia
SNPc Substantia nigra pars compacta
SPMS Secondary progressive multiple sclerosis
SSADH Succinate semialdehyde dehydrogenase
STN Subthalamic nuclei
TBI Traumatic brain injury
TGB Tiagabine
THDOC Tetrahydrodeoxycorticosterone
THIP Tetrahydroisooxazolopyridinol
VGB Vigabatrin
VLPO Ventrolateral preoptic nuclei
VTA Ventral tegmental area
8.1 Introduction
In the brain, neuronal excitation and inhibition is responsible for the basis of
information transfer within the CNS. Amino acid neurotransmitters are the
messengers which exert excitatory and inhibitory actions, which include glutamate
and GABA, respectively. The balance between these two neurotransmitters ensures
normal functioning of neurons showing rhythmic activity. GABA is the main
inhibitory neurotransmitter in the brain and is made up of amino acid sequences.
The primary action of GABA is exerted by binding to the postsynaptic receptor,
which stimulates the Cl ion channels, opens Cl ions channels resulting in hyper-
polarization of the cell, and thus exerts the inhibitory response, i.e., conductance of
action potential is completely blocked. Clinically, the significance of GABA has not
been estimated yet, but mainly it controls the transmission of neurotransmitters in the
synapses and excitability of the neurons within the CNS. The ubiquitous distribution
of GABA in the brain helps to exhibit a variety of physiological functions including
244 S. Kaur et al.
calmness, sleep, motor functions, cognition, and memory like behavioral phenome-
non (Wu and Sun 2015). No doubt, it has an inhibitory control over mammalian brain
but in neonatal brain it also exhibits some excitatory functions. Here, it participates in
a variety of functions including neurogenesis, synapse formation, and synaptic plas-
ticity. This indicates that GABA is well implicated in normal neural development and
physiology of the brain. The inhibitory nature of GABA helps to execute over the
excitatory neurons for maintaining the homeostasis between GABA and glutamate
which is known as GABA/glutamate cycle. Any asymmetry in this balance creates the
pathological situations associated with different disorders like epilepsy, schizophrenia,
anxiety, dementia, and motor disorders like Parkinson’s disease. Such pathological
associations make GABAergic dysfunction as a major target for significant therapeutic
approach in CNS disorders (Wu and Sun 2015). GABA signaling modulation or
enhancing GABAergic inhibition is the way for the treatment of many pharmacologic
drugs in various pathological situations.
GABA is present in different parts of the mammalian body, but its various roles
are still unknown. Primarily it shows inhibitory responses in the CNS as well as in
the spinal cord. Usually, the main function of the beta cells of the pancreas is to
produce insulin but is also able to produce GABA. Thus GABA produced from the
beta cell can stimulate the growth of beta cells, formation of beta cells from alpha
cells, and inhibiting alpha cells. Within the body, the other parts contain low
amounts of GABA, though the proper function and relevant clinical significance
of this remain unknown, but it mainly counterbalances the excitatory responses.
Firing of the neuronal cell is too fast, in the absence of GABA. When GABA
activity is significantly decreased in the CNS, various disorders like anxiety,
convulsions, panic attacks, cognitive dysfunction, Parkinson’s disease, drug addic-
tion, etc. occur due to excitatory discharge. Transmission of nerve impulses is
significantly hindered from one neuron to another by GABA, due to inhibitory
responses; it has calming or quieting effect. In the brain, the activity of growth
hormone, synthesis of protein, and plasma concentration are improved by GABA,
but diminishes small airway-derived lung adenocarcinoma. Other responses include
hypotensive, tranquilizing, diuretic, and antidiabetic effects.
Inhibitory neurotransmission is a complex process but it can be achieved through
the activation of different types of GABA receptors. Historically only two types of
GABA receptors (i.e., GABAA and GABAB) were identified, but now GABA
receptors are classified into three types (GABAA, GABAB, GABAC) based on phar-
macological and electrophysiological properties. Recently, GABAA and GABAC are
classified as ligand-gated chloride channel receptors and GABAB receptors are
known as metabotropic, i.e., G-protein-coupled receptors.
The therapeutic reliability of neurotransmitter GABA could be analyzed from
the success of benzodiazepines (BZD). BZDs are a class of drugs which act by
interacting with GABA receptors and utilized as the most commonly prescribed
drugs clinically. BZDs are the positive allosteric modulator of GABAA receptor to
potentiate the inhibitory neurotransmission derived calming effect in the brain
(Griffin et al. 2013). The drugs under BZDs include diazepam, flurazepam, alprazo-
lam, chlordiazepoxide, etc. which are commonly prescribed for anxiety, insomnia,
8 Pharmacology of GABA and Its Receptors 245
In 1883, it was known only as a plant and microbial metabolic product. There are
evidences which establish that in the mammalian CNS, γ-aminobutyric acid
(GABA) is the most important inhibitory neurotransmitter which was fully accepted
at the end of the 1960s/early 1970s by its role. In 1910, it was shown to be present in
biological tissues but at that time its presence in the mammalian CNS was not
confirmed. Only after 1950, i.e., 40 years later, the presence of free amino acid
was identified in the brain that arose the neurochemical significance of GABA.
During the ensuing decade, how neuronal activity is affected by GABA and related
compounds has been reported to define its role within the CNS.
Additionally, the role of excitatory and inhibitory activities of GABA in crusta-
cean is defined by concurrent findings of Kuffler (1954) (Jijón-Lorenzo et al. 2018).
The initial finding related to GABA was muted. After 5 years of discovery by
Roberts and Frankel, only two articles related to GABA activity within the brain
were reported in the Journal of Biological Chemistry and PubMed had only four
articles related to GABA activity. Nevertheless, in those days due to the lack of
methods that define its presence and function, research community remained silent.
The inkling properties of GABA were not shown by anyone in the brain. In 1957,
researchers in Canada confirmed the activity of GABA due to the inhibitory activity
of an unknown compound on crayfish neuron. GABA transporters were studied by
Baruch Kanner of Hebrew University, Hadassah Medical School in Israel, who is a
member of the Journal of Biological Chemistry editorial board.
Kanner said, “GABA receptors have an inhibitory input which is the major
inhibitory action within the brain.” Roberts and Frankel formed the basis and provided
evidence; then only it was clear about how neurotransmitters control the brain activity.
Roberts analyzed various extracts and reported the existence of a free amine com-
pound by migration of ninhydrin reactive compounds on chromatograms. This amino
acid is accumulated in higher concentrations in the brain than in other tissues.
In 1987, Eric A Barnard in Cambridge (UK) who is a molecular biologist and
Peter H Seeburg at Gene tech in San Francisco (CA, USA) succeeded to define
GABAA receptor, which is then classified as ligand-gated ion channels. After a
decade, airway epithelium containing GABAergic system which is excitatory was
described in 2007. On exposure to allergens the system becomes activated
and participates in the mechanisms of asthma. In the testis and eye lens GABAergic
system is present as shown in Table 8.1.
246 S. Kaur et al.
The synthesis, storage, release, reuptake, and metabolism of GABA occur due to
signaling of the GABA system. Both ionotropic and metabotropic receptors
expressed in the plasma membranes of cells and GABA exert their action through
these receptors (Olsen and Sieghart 2009). GABA is formed by various metabolic
pathways, also called the GABA shunt, and operates via closed loop process. The
purpose of this pathway is to produce and conserve the supply of GABA. Brain
regions contain high amount of GABA and the amount is 1000 times greater than
other monoamine neurotransmitters. The endogenous synthesis of GABA proceeds
via α- decarboxylation of L-glutamate by action of the enzyme glutamic acid
decarboxylase (GAD) as shown in Fig. 8.1. The functioning of GAD depends
upon the presence of cofactor pyridoxal-50 phosphate to catalyze the single-step
irreversible reaction for production of GABA (Roth and Draguhn 2012). Here, GAD
exists in two forms; GAD 65 (65 kDa) was found to reside in axons and
synaptosomes along with plasma membrane whereas GAD67 (67 kDa) was found
to be localized in the cytosol of neuronal cells. These both mediate the synthesis of
GABA by vesicular mechanism and cytoplasmic production, respectively.
On production, GABA gets recruited to synaptic vesicles via vesicular GABA
transporter (vGAT), which releases GABA in synapse on membrane depolarization
of neurons. After the release, GABA interacts with its receptors for mediating its
action. The small amount of GABA release is re-uptaken by membrane-bound
GABA transporters (GATs), localized on neurons and astrocytes. The reuptake of
GABA is temperature and ion dependent. The GABA transporters are usually
GABA/Na+/Cl symporters that constitute four different types including GABA
transporter 1 (GAT1), GABA transporter 2 (GAT2), GABA transporter 3 (GAT3),
and the betaine-GABA transporter (BGT1). These transporters are engaged over
synapses to clear GABA from the synaptic cleft (Roth and Draguhn 2012). Further,
hetase
248
Ca2+
chann
el
Astrocyte
GAT
K+ channel
GABAAR
BR
GABA
Synaptic receptor Extrasynaptic receptor
Postsynaptic Terminal
GABA
Succinic semialdehyde
Succinic acid
Neurotransmitter has the ability to affect the synaptic transmission by binding to the
postsynaptic receptors. In the past, based on different structural and pharmacological
classes, GABAergic receptors were divided into two distinct classes, i.e., GABAA
and GABAB. But now the third type of GABA is also recognized, i.e., GABAC. At
the end of 1980, the structure elucidation of the GABAA receptor was done after the
clarification of the receptor subunits; it is the member of the superfamily to which
various other types of receptors are included such as nicotinic acetylcholine and
glycine. GABAρ receptor which was initially known as GABAC receptor is a
subclass of GABAA receptor and is found in the retina (Johnston et al. 2003).
The GABAA receptor is the ligand-gated ion channel (anion selective) that
induces inhibitory responses and is made up of different subunits of 8 subfamilies
250 S. Kaur et al.
α (1–6), β (1–4), γ (1–3), δ, ε, θ, π, and ρ (1–3) (Sigel and Steinmann 2012) as shown
in Fig. 8.3.
Various combinations are possible when different 19 subunits are combined in
the pentameric structure. However, the nerve cells contain around 25–30 GABAA
receptor combinations (Birnir and Korpi 2007; Olsen and Sieghart 2009). Most
common subunits are α, β, and γ which assemble to form a sequence of unique
functionality, while δ, ε, and ρ remains less common and elusive for its functions
(Sigel and Steinmann 2012). The most common ones are enlisted as α1β2, α3β3γ2,
and α2β3γ2. The available research data provides evidence regarding the role and
precise locations of GABA subunits. Immunocytochemistry studies reveal the wide
distribution of α1, β1, β2, β3, and γ2 throughout the brain (Olsen and Sieghart 2009).
The expression of β2 is specifically confined to the cerebellum, thalamus, hypothal-
amus, olfactory bulb, amygdala, and midbrain, whereas β3 is highly localized in
cerebellum and midbrain regions like substantia nigra pars compacta and the ventral
tegmental area. The expression of α2, α3, α4, α5, and α6 remains more region
specific as can be studied from an example of α5 which is highly localized in
olfactory bulb, hippocampus, and hypothalamus (Rudolph and Möhler 2014), and
the α4 subunit which is highly localized in the thalamus, striatum, hippocampus, and
outer cortex. Apart from specific regional findings, it is neuron specificity that
reveals the presence of GABAergic subunits using immunofluorescence staining.
The result of these studies postulates the pallidum neuron as α1 specific while
brainstem as α1 and α3 specific. Moreover, their widespread distribution of seroto-
ninergic neurons inside the raphe nuclei has α1 subunit of GABA while GABAergic
neurons here express specificity for α1 and α3 subunits (Wu and Sun 2015). This
way the widespread distribution of subunits and localized expression all over the
CNS performs a number of various actions as shown in Table 8.2.
The GABA network is highly distributed in the CNS and plays a role in neuronal
excitability. Different brain regions contain different expression of the receptor and
distribution based on the function of the subunit as shown in Table 8.3.
8 Pharmacology of GABA and Its Receptors 251
Table 8.3 Different subunits in different regions with concentrations of GABAA receptors
Concentration
Subunits (%) Regions
α1β2γ2 60 In most brain areas and it is localized in interneuron in
hippocampus and cortex Purkinje cells
α2β2/ 15–20 Forebrain especially hippocampus/striatum
3γ2
α3βnγ2/ 10–15 Primarily in cortex
γ3
α2βnγ1 8 Bergman glia
α4βnδ 5 Thalamus and hippocampal dentate gyrus
α4βγ2 5 Thalamus and hippocampal dentate gyrus
α4βnγ 5 Hippocampal pyramidal cells, deep cortical layers, amygdala,
olfactory bulb, hypothalamus, superior colliculus, spinal
trigeminal nucleus
α5β3γ2/ 4 Hippocampus, cortex, and olfactory bulb
γ3
α5β2γ2 5 Hippocampus
α6β2/ 5 Cerebellum and dorsal cochlear nucleus
3γ2
α6βγ2 2 Cerebellar granule cell
α6βnδ 2–3 Cerebellar granule cell
ion conductance of the receptor and fate of GABA are affected by intracellular loop
because it contains the binding site of intracellular proteins and phosphorylation
proteins (Olsen and Sieghart 2009) as shown in Fig. 8.4.
There are various binding sites located in the complex but the GABA binding site
is between the interface of α and β subunits and other binding sites can modulate the
action of GABA. Various drugs such as benzodiazepines, barbiturates,
neurosteroids, and ethanol (Sieghart et al. 2012) positively modulate the GABAA
receptors where as GABAA receptor blocked by bicuculline and picrotoxin
(Semyanov 2002) as shown in Fig. 8.5. Different brain regions contain a combina-
tion of various subunits of GABAA receptor performing different functions at the
same time. Each class of units has different pharmacological and electrical properties
and performs specific functions.
On the other side, the metabotropic receptor (GABAB) belongs to G-protein-
coupled receptors (GPCRs) or metabotropic receptors for GABA and acts by
coupling with G-proteins to mediate the intracellular signaling pathway. The
8 Pharmacology of GABA and Its Receptors 253
GABAB receptors were determined by Norman Bowery and their team in 1981 when
they are distributed in the CNS (Blackburn 2010). The GABAB receptors are
activated by GABA which is the main inhibitory neurotransmitter present in
the CNS. The GABAB receptors presynaptically inhibit voltage-activated Ca2+
channel to reduce the release of GABA and other neurotransmitters. On the other
side, the GABAB receptor postsynaptically activates Kir3 channels and inhibits the
neuronal activity by generating hyperpolarizing postsynaptic potentials, which over-
all minimize the excitatory neurotransmission of GABAB receptors by interacting
with their effectors through Gα and Gβγ subunits of the G protein; the first effector of
GABAB remains adenylate cyclase (AC) via activation of an inhibitory subunit
(Gαis/Gα/Gβγ) that further inhibits voltage-dependent Ca2+ channels to limit the
neurotransmitter release (Pin and Prézeau 2007). Here, GABAB receptors act as
autoreceptors which therefore will inhibit the release of GABA and also
the excitability of neurons. The GABAB receptors stimulate the opening of K+
channels which brings the neuron closer to the equilibrium potential of K+ thereby
reducing the frequency of action potential and the release of neurotransmitters.
Hence, GABAB receptors are inhibitory receptors. These receptors also reduce the
activity of adenylyl cyclase and Ca2+ channels (Orts-Del’Immagine and Pugh 2018).
Another important mechanistic regulation of GABAB involves activating the Kir3
channels which maintain slow inhibitory postsynaptic potentials (IPSPs) by inducing
efflux of K+ ions for hyperpolarization of the membrane (Pin and Prézeau 2007) as
shown in Fig. 8.6.
Further, GABAB is of two subtypes: GABAB1 which activates neurons and
GABAB2 that mediates specific sequential signaling. The GABAB1 further consists
254 S. Kaur et al.
of GABAB1α and GABAB1β like subtypes; here the GABAB1α carries a specific
domain named “sushi,” to control the physiological release of glutamate at presyn-
aptic terminals. The GABAB1α is encoded by the GABAR1 gene. But, GABAB1β
receptors are located on postsynaptic terminals, encoded by GABBR2 gene and
cause inhibition there. The GABAB1α and GABAB1β are isoforms and combine with
GABA to form a functional GABAB(1a,2) and GABAB(1b,2).
Another receptor for GABA includes GABAC which shows similarity to GABAA
receptor. The GABAC receptors were first described in interneurons of the spinal
cord. It remains ionotropic in nature and their pentameric chloride channels are com-
posed only of ρ (1–3) subunits. This receptor is found to be expressed in the retina,
observed by Miledi from bovine retina in Xenopus oocytes, and unlike GABAA it
remains insensitive to bicuculline and baclofen like compounds (Johnston et al.
2003). GABA has high sensitivity for GABAC receptors. The pharmacological
profile of GABAC receptors is different from GABAA and GABAB receptors. In
rats, tiger salamander, and hybrid bass, GABAC receptors were found on bipolar
cells (Du and Yang 2000). It is evidenced that GABAC receptors are composed of ρ
subunits (ρ1,2 in humans; ρ1,2 in rat; ρ2,3 in rat; ρ1,2 in chicken) (Zhang et al. 2001)
as shown in Fig. 8.7 and Table 8.4.
8 Pharmacology of GABA and Its Receptors 255
GABA receptors exert inhibitory responses after binding to the neurotransmitter, i.e.,
gamma-aminobutyric acid (GABA).
GABAA receptors are ligand-gated ion channels which are pentameric proteins in
nature and constitute 5 subunits belonging to different families (α1–6, β1–3, γ1–3, δ,
π, ε, ρ, θ). They contain an integral chloride channel and hyperpolarize the receptor,
and have modulatory sites for barbiturates, neurosteroids, ethanol, and
benzodiazepines.
GABAB receptors are metabotropic in nature, i.e., G-protein-coupled receptors
and exist as heterodimers of GABAB1 and GABAB2 subunits. The GABAB1 subunit
is able to bind with GABA, and the GABAB2 subunit shows interactions as shown in
Table 8.5.
8.6.1 Sleep
Sleep in human beings and animals has been partitioned into rapid eye movement
sleep (REMS) and non-REMS (NREMS). REMS keeps up the house-keeping
function of the brain and its loss influences the vast majority of the psycho-physical
physiological processes (Yadav et al. 2019). Another report proposes the sleep
promoting neurons (SPNs) incorporate GABAergic neurons in the ventrolateral
preoptic nuclei (VLPO)/intermediate nuclei, middle preoptic nuclei (MnPO), and
brainstem parafacial zone (Ma et al. 2019; Anaclet and Fuller 2017).
Outstanding among other best-studied co-transmission system is the co-release of
glutamate by primary GABAergic/glycinergic neuron terminals in the medial
nucleus of the trapezoid body (MNTB). For instance, starburst amacrine cells
(SACs) in the retina discharge GABA through vesicles in a Ca2+ dependent process
(Lee et al. 2010). This enables few SACs to encode both movement and course
sensitivities using GABA signaling, respectively. Glutamate is co-released from
dopaminergic neurons in the ventral tegmental zone (VTA) (Chuhma et al. 2009).
Dopamine acts upon a moderate timescale by binding to G-protein-coupled
receptors, while glutamate acts upon a fast timescale when bound to ionotropic
glutamate receptors and conveys temporarily exact signals. Glutamate co-release is
helpful for exact prediction error signals, allowing the reward to be encoded in the
firing rates of dopaminergic neurons and mediating dopamine-dependent behaviors
(Mingote et al. 2017).
GABA also appears to play a role in female reproductive functions, perhaps because
hormonal cycles may synchronize with the body clock. GABA shows an increase in
256 S. Kaur et al.
Table 8.5 Selective agonist and antagonist of GABAA, GABAB, GABAC receptors
Name of compound Description
GABAA
Muscimol Competitive GABAA receptor agonist
GABA Endogenous agonist
Isoguvacine hydrochloride Selective GABAA agonist
L-838,417 GABAA partial agonist; displays subtype selectivity
MK 0343 GABAA partial agonist; displays subtype selectivity
Muscimol Potent GABAA agonist; also GABAA-ρ partial agonist
TACA GABAA agonist; also GABA-T substrate and GABA uptake
inhibitor
TCS 1205 GABAA agonist; displays subtype selectivity
THIP hydrochloride GABAA agonist
ZAPA sulfate “Low affinity” GABAA agonist; also GABAA-ρ antagonist
SR 95531 hydrobromide Potent, selective, competitive GABAA receptor antagonist
(Gabazine)
(-)-Bicucullinemethiodide Competitive GABAA receptor antagonist
Picrotoxin Noncompetitive GABAA receptor antagonist/glycine receptor
inhibitor
Flumazenil GABAA receptor antagonist
MRK 016 α5-selective GABAA inverse agonist
TB 21007 α5-selective GABAA inverse agonist
Furosemide GABAA antagonist; also Na+/2Cl-/K+ cotransporter blocker
PHP 501 trifluoroacetate Potent GABAA antagonist
Picrotoxin GABAA antagonist
SCS Potent GABAA antagonist; β1-subunit selective
SR 95531 hydrobromide Competitive and selective GABAA antagonist
Suramin hexasodium salt Competitive α1β2γ2 GABAA antagonist; also nonselective P2
antagonist
GABAB
Baclofen Selective GABAB receptor agonist
SKF 97541 Potent GABAB receptor agonist
(RS)-Baclofen Selective GABAB agonist
GABA Endogenous agonist
SKF 97541 Highly potent GABAB agonist; also GABAA-ρ antagonist
CGP 55845 Potent, selective GABAB receptor antagonist
Saclofen Selective, competitive GABAB receptor antagonist
SCH 50911 Selective, competitive GABAB receptor antagonist
CGP 35348 Selective GABAB antagonist; brain penetrant
CGP 36216 hydrochloride GABAB antagonist; displays activity at presynaptic receptors
2-Hydroxysaclofen Selective GABAB antagonist; more potent than Saclofen
Phaclofen Selective GABAB antagonist
SCH 50911 Selective and competitive GABAB antagonist
GABAA-ρ receptors/GABAC
GABA Endogenous agonist
(continued)
8 Pharmacology of GABA and Its Receptors 257
the levels of luteinizing hormone, estrogen, and progesterone while decrease in the
level of FSH. GnRH neurons express both GABA and GABA receptors and receive
GABAergic input that expresses estrogen receptors; therefore GABA has been
implicated as a major player in the regulation of GnRH neuron activity and secretion
(Maffucci and Gore 2009).
intracellular cascade culminating in the synthesis of new proteins, which are utilized
for the synaptic changes required to stabilize the new, reactivated, or inhibitory
memory trace (Makkar et al. 2012).
In the mammalian central nervous system, GABA is the main inhibitory neurotrans-
mitter. GABAA and GABAB receptors are activated by aminobutyric acid in
neurocytes, which promote the dilation of blood vessels and also reduce blood
pressure (Ma et al. 2015). GABA reduces the renal sympathetic nerve secretion in
parallel with the fall in blood pressure. Several studies reveal that stimulation of the
GABA receptor can centrally reduce sympathetic nerve activity, causing a drop in
blood pressure. Although the physiological significance and location of these
receptors are unknown, the pharmacological manipulations of these receptors can
cause extreme functional changes and involve GABA as a possible central
modulator of cardiovascular control.
8.6.5 Heart
GABA slows heart rate by activating GABA receptor. GABA inhibits the central
norepinephrine neurotransmitter system and produces the tension booster effect and
may increase the heart rate. Experimental study suggests that muscimol decreased
heart rate in insulated heart experiments and may represent an effect on intrinsic
cardiac activity (Bentzen and Grunnet 2011). Bicuculline, a GABA antagonist,
shows a negative chronotropic effect which can be described by the excitatory
actions of vagal neurons. Reports suggest that GABA has an excitatory effect in
early development due to high chlorine intracellular concentrations. The change has
inhibitory GABA phenotype due to the increased aspect of the cotransporter KCC2
chlorine output (Ben-Ari 2002). Lin et al. reported that there is a correlation between
the reduction in HR and temperature and state that the temperature could influence
ion channels, hence extending the duration of a cardiac tissue action potential, with a
greater effect on atrial tissue (Lin et al. 2014). Rana et al. reported a decrease in HR
with exposure to caffeine and caused cardiac arrest in some cases. The reports
suggested that the negative chronotropic effects could be due to the presence of
adenosine receptors, which modify the activity of potassium channels, sustain
cardiomyocyte membrane hyperpolarization and therefore reduce heart rate (Rana
et al. 2010).
8 Pharmacology of GABA and Its Receptors 259
Fig. 8.8 Mechanism of action of GABA and BZD on nerve cells in the brain
260 S. Kaur et al.
Inhibitory synapses are the most abundant synapses in the CNS and include neuro-
transmitter GABA. Aspects of health, especially emotional and physical stability, are
affected by GABA. During locomotion specifically, it relaxes the body muscles and
foraging and helps in the process of defecation by contracting the enteric muscle.
The important functions of this neurotransmitter are included in various natural
processes and pathological processes as it is required for basic motor function.
Various types of neurological and psychiatric disorders including anxiety, depres-
sion, insomnia, and epilepsy occurred due to the deficiency of GABA and also due to
the obstruction of electrical impulses. GABA has been shown to affect many
biological functions in the brain, e.g., cognition, learning, emotions, locomotion,
circadian rhythms, and sleep. Furthermore, GABA also has roles in cellular events
such as differentiation, proliferation, migration, axonal growth, synapse formation,
and neuronal death (Kilb 2012; Birnir and Korpi 2007).
Pharmacological actions are responsible for the GABAA effects including
excitability of the neuron (Carver and Reddy 2013), anxiety modulation (Nuss
2015), behavior (Caldji et al. 2004), circadian rhythms (Albus et al. 2005), and
learning and memory abilities (Chapouthier and Venault 2002).
receptors, known as GABAC. Initially, the receptor comprised only three subunits
(ρ1, ρ2, and ρ3), but later it was revealed that five (ρ4 and ρ5) subunits form the
ionotropic GABAC receptors necessary to form a functional channel (Zhang et al.
2001). Also, the brain regions contain the expression of ρ subunits with function in
the visual pathways (Lukasiewicz et al. 2004) as well as in the local GABA circuit of
developing visual cortex (Morales et al. 2002), so expression is not only restricted to
the retina and does not indicate a separate ionotropic receptor subfamily. It was
confirmed that the human retina contains ρ2 subunits with significant abundance
(Naffaa et al. 2017) and other regions of the brain such as hippocampus, cerebellum,
and pituitary were detected with lower expression levels (López-Chávez et al. 2005).
The sensitivity of GABA to GABAC receptors is more as compared to GABAA
and GABAB receptors. Ionotropic GABAC receptors desensitization and opening
time of channel are also longer than in the other receptors. Excellent models are
available to characterize GABAc receptors in the retina, i.e., rod-driven horizontal
cells and availability of GABAc receptors on other types of retinal neurons.
The influence of GABAC receptors on sleep-waking processes is limited.
GABAA-modulatory drugs such as benzodiazepines, barbiturates, and neurosteroids
do not affect GABAC receptors. GABAC receptors unlike the GABAA receptors
show different electrophysiological responses and is additionally sensitive to the
physiological agonist while the Hill slopes are steeper reflecting the presence of five
ligands binding site whereas two sites appear to be present on the GABAA receptor.
GABAA receptors (such as SR95531 and hydrastine) or GABAB receptors (such as
phaclofen and saclofen) antagonists are not sensitive to GABAC receptors on retinal
neurons. Detailed studies indicated that GABAC receptor binding preferences
depend on different conformation of GABA molecule.
Recently, a novel target for analgesia is ρ2 receptors subunit implicated in pain
perception found presynaptically in the spinal dorsal horn (Tadavarty et al. 2015).
Sleep-waking behavior of rats demonstrated by behavioral pharmacological studies
(Arnaud et al. 2001), learning and memory in chicks and rats (Chebib et al. 2009),
the inhibitory modulation of the olfactory bulb (Chen et al. 2007) involves ρ1
receptors.
clinical studies under AD research (Li et al. 2016). This way evidence-based studies
over a number of pathogenic mechanisms made implicit different agonists. Inverse
agonists and antagonists for GABA for therapeutic approach in AD are shown in
Table 8.6.
Not only this, non-motor symptoms are believed to be brought out by GABAergic
dysfunction. Here, the 80% of cases of PD reports for abnormal olfactory functions.
The distorted olfactory function in PD is associated with the loss of dopaminergic
neurons in olfactory bulbs and olfactory nuclei which depends upon glial cell-
derived neurotrophic factors (GDNF) for survival. The availability of GDNF
depends upon neuro-glial interactions effected by GABA/Ca2+ mechanism in both
midbrain and olfactory nuclei. During normal conditions, the physiological release
of GABA causes hyperpolarization of neurons for long-term inhibition of synaptic
transmission. This hyperpolarization phase blocks the calcium channel and protects
neurons from excitotoxicity (Błaszczyk 2016). The removal of calcium from
mitochondria and cytoplasm requires a high amount of energy; therefore neuronal
cells in the midbrain have energy requirements to cope with calcium overload. On
the other side, calcium overloads lead to oxidative stress-mediated dysfunction of
mitochondria. This way subsequent decline in ATP production, increase in oxidative
stress, and calcium load result in mitochondrial dysfunction-mediated neuronal loss
in SnPc in PD (Błaszczyk 2016). The excitotoxicity mediated initial release of
GABA from astrocytic neurons but on exceeding the limits density of synaptic
GABA receptors gets decreased which leads to an increase of GABA in synaptic
cleft and initiate self-mediates apoptosis in neuronal cell. Furthermore, the defect in
GABA-glutamate cycle (neuron-astrocyte interaction) for GABA recycling could
also result in neuronal damage as shown in Table 8.7.
270 S. Kaur et al.
Among different receptors for GABA, the receptor GABAA remains a potential
target for therapeutics in HD. The distribution of GABAA in striatum mediates tonic
(α5 by α 1–3, δ or by γ 2 forming extrasynaptic receptor) and phasic inhibition (α1,
α2, α3 in combination with β and γ2) via its different subunits (Waldvogel and Faull
2015). Here, α1 facilitates the fast inhibitory current and α2 subunit gets altered and
hence confirms the decrease in GABAA in HD brain. The α1 participates in the
co-release of GABA and dopamine in medium spiny neurons of the striatum and its
decreased expression contributes to motor symptoms of HD.
Further, there is a spatial distribution of GABAergic interneurons (IN) in HD,
different classes of IN including fast-spiking INs (FSIN) disposing parvalbumin,
persistent low-threshold spiking INs (PLTS) expressing somatostatin or nNOS, and
INs expressing calretinin. Specific degeneration of fast-spiking neurons specifically
contributes to HD (Du et al. 2017). Moreover, the role of cholinergic IN remains
essential to discuss as they are widely distributed in the striatum which bears
localization of GABA receptors. The increase in α3 subunits of GABAA receptor
on cholinergic IN mediates inhibition over acetylcholine release in HD. Decrease in
Tonic inhibition also a prominent pathogenic mechanism in HD, which is confirmed
by a decrease in tonic inhibition regulatory subunits like α5 and δ (Allen et al. 2009).
Moreover, the reduced tonic current mediates the movement and psychiatric
symptoms in patients affected with HD.
The GABAergic neurons remain major inhibitory control above overexcited GPe,
so it is essential to discuss the subunits that are expressed there. The α1, β2, and
γ2 subunits get reduced along with decreased expression of vesicular GABA
transporter in GPe, which is also reported in HD. Moreover, the subunits like α2,
α3, and γ1 remain expressed in GPe, which is reported to get altered in the HD brain
(Du et al. 2017). So, in this way specific agonists and antagonists for different
subunits could be more reliable towards therapeutics in HD.
272 S. Kaur et al.
further reliability. This way several different subunits-specific molecules are under
investigation for lesser side effects with improved anxiolytic profile for therapeutic
benefits.
Not only GABAA but GABAB also provides some remarkable therapeutic effects
in anxiety. There are some endogenous steroids which also show specific binding
towards the GABAA for anxiolytic activity with lesser side effects than BZDs.
Animal studies revealed the anxiolytic effects of different neurosteroids including
pregnenolone, dehydroepiandrosterone, and progesterone even at low
concentrations (Nuss 2015). Etifoxine, which is a small structurally unrelated mole-
cule to BZDs, promotes neurosteroids synthesis in the brain that shows anxiolytic
effects in both animals and humans. The anxiolytic effects of etifoxine are
contributed by positive allosteric modulating effect on GABAA by binding to β
subunit specifically β2 and β3 sites. Therefore, GABAergic system potentially
participates in the regulation of anxiety and could be a novel approach for future
studies.
evidenced the alterations in GABA levels to confirm its role in altered activity of
the brain. Decreased activity of GABAergic interneuron observed in PFC along with
reduced quantity of GAT1 and GAD67. The reduction in GABAergic signaling in
PFC may contribute to cognitive, emotional, and enhanced dopaminergic function
like abnormalities of SCZ patients (Nakazawa et al. 2012). The justification behind
this associates with dysfunctioning of mesostriatal dopaminergic neurons in SCZ.
Normally, the dopaminergic projections of mesostrial pathway give input to PFC
GABAergic neurons, and this control becomes mature during younger age with rise
in cortical activity. But any genetic or environmental risk may hinder this control by
causing dysfunction in mesostriatal dopaminergic pathway which reduces GABA in
SCZ (Selten et al. 2016). However, a decrease in dendritic spines and loss of
pyramidal neurons also contribute to a decrease in GABA in SCZ.
Alteration in GABA levels contributes to diverse symptoms of SCZ including
synaptic plasticity, memory, execution, and psychosis. The loss of parvalbumin
GABAergic interneurons in the hippocampus and cortex may contribute to executive
and cognitive symptoms of SCZ (Lewis et al. 2012). One report suggests that
a decrease in GABAergic neurotransmission in the hippocampus contributes
to deficits in working memory in SCZ (Gao and Penzes 2015). Moreover, psychosis
is considered as the positive symptom of schizophrenia that triggers by dysfunction
of the dopaminergic system in the schizophrenic brain (Kesby et al. 2018). The
hyperexcitation of dopamine (D1) in the striatum is reported to crucially control
cortical activity. As if the cortical interneuron gets inhibited, striatum dopamine
neurons get overexcited that may result in schizophrenic symptoms. The debatable
possible mechanism is that it may decrease in cortical GABAergic interneurons
activity that inhibits ventral tegmental area to increase nucleus accumbens dopamine
activity (Nguyen et al. 2014).
Accordingly, a number of drugs targeting dopamine are going to be evaluated for
their beneficial role in SCZ. Direct infusion of GABA agonist muscimol in the hip-
pocampus is shown to mitigate the memory deficits caused by phencyclidine (PCP)
treatment (Riordan et al. 2017). The hormonal drug estradiol rescues memory in
SCZ by upregulating the expression of GABA in the hippocampus. Moreover, a new
compound named as imidazenil modulates GABAergic neurotransmission for bene-
ficial effects in SCZ. It is highly potent partial GABA agonist with anticonvulsant
and anxiolytic property without any side effect of sedation (Guidotti et al. 2005).
Moreover, recent studies are also assessing the therapeutic efficacy of GABA
targeting drugs via their subunit selectivity. The different subunits of GABAA
including α5, α-2/3, and γ2 are ongoing major therapeutic targets in SCZ. The
selective α5 inverse agonists and α-2/3 agonists are under preclinical studies for
their therapeutic benefits over impaired cognitive and executive functions of SCZ
(Charych et al. 2009). The γ2 subunit of GABAA is reported to participate in
physical interaction with D5 receptor of dopamine which may initiate the symptoms
of SCZ (Vinkers et al. 2010) as shown in Table 8.11.
278 S. Kaur et al.
Traumatic brain injury (TBI) is a neurologic disorder resulting from head injury that
leads to temporary or permanent impairment of the structure and function of
the brain. It could occur due to violent blow, injury, or aggressive shakiness to the
head which produces rapid accelerated or decelerated impact on the brain that leads
to structural damage, functional dysfunction, or neurological deficits leading to
the death of patients. The most common TBI-associated cases are prevalent in traffic
accidents, military, sports, violence, construction sites, industrials, etc. (Madikians
and Giza 2006). These cases also occur in infants when sudden shake to babies cause
a violent impact on their heads. The common pathogenic features of TBI involve the
disturbance of the protective endogenous antioxidant system along with altered
mitochondrial function, excitotoxic damage, and cerebral ischemia. Excitotoxicity
is considered as the major pathological event occurring in TBI brain. It is associated
with an imbalance in between the GABA/Glutamate ratio that raises susceptibility of
neurons towards cell death (Yi and Hazell 2006). The glutamate is the major
neurotransmitter present in pyramidal neurons localized in midbrain, hypothalamus,
cortex, and hippocampus, while GABA interneurons are localized in cortical and
thalamocortical regions to regulate motor functions, attention, and memory. The
adequate balance of GABA/Glutamate is necessary for neuron survival; any shift to
it results in cerebral ischemia, TBI, and other neurodegenerative disorders.
During focal TBI, penetrating injury causes local swelling and ischemia that may
irreversibly damage localized tissue through excitotoxicity. One report suggested
that posttraumatic epilepsy is caused by an imbalance in GABA and glutamate. The
imbalance is reported to result from the loss of parvalbumin-positive GABA neurons
(Guerriero et al. 2015). Moreover, brain studies of concussion-affected athletes
reported that there is an increase in levels of GABAB after injury. It is believed to
be proceeded to counteract the excitotoxicity-induced neuronal damage for survival.
Further, synthesis of GABA is occurring from glutamic acid decarboxylase and its
deficiency results in a decrease in GABA levels in the TBI brain. Some evidence has
also contraindicatory reports that speculated that abnormal synaptic activity in
the TBI brain results from a decrease in glutaminergic neurotransmission and
8 Pharmacology of GABA and Its Receptors 279
Most of the cases of epilepsy are idiopathic while some may be secondary due to
trauma/intracranial tumor/cerebral ischemia. GABA is present relatively in high
concentration in the mammalian brain which is further catalyzed by GABA amino-
transferase into glutamate, i.e., an excitatory neurotransmitter responsible for
the occurrence of epileptic seizures (Khazipov 2016; Rao and Zhang 2011),
Alzheimer’s disease (Solas et al. 2015), Huntington’s disease (Byrne and Wild
2016), Parkinson’s disease (Silverman 2012), tardive dyskinesia, anxiety, schizo-
phrenia, and other behavioral disorders (Gajcy et al. 2010). The explanation for
seizures is the inhibition of glutamate decarboxylase, a synthesizing enzyme of
GABA by lowering brain GABA levels.
Experimental and clinical investigations demonstrate that GABA has a significant
role in the mechanism and treatment of epilepsy: (1) Abnormalities of GABAergic
function have been seen in genetic and acquired animal models of epilepsy;
(2) Reductions of GABA-mediated inhibition, activity of glutamate decarboxylase,
binding to GABAA and benzodiazepine sites, GABA in cerebrospinal liquid and
brain tissue, and GABA detected during microdialysis studies have been accounted
for investigations of human epileptic brain tissue; (3) GABA agonists suppress
seizures, and GABA antagonist produces seizures; (4) Drugs that inhibit GABA
synthesis cause seizures; and (5) Benzodiazepines and barbiturates work by enhanc-
ing GABA-mediated inhibition. Finally, drugs that increase synaptic GABA are
potent anticonvulsants. Two recently developed antiepileptic drugs (AEDs),
vigabatrin (VGB) and tiagabine (TGB), are examples of such agents. However,
their mechanisms of action are quite different (VGB is an irreversible suicide
inhibitor of GABA transaminase, though TGB blocks GABA reuptake into neurons
and glia), which may represent observed differences in drug side effect profile (van
Vliet et al. 2018; Löscher et al. 2013; Treiman 2001).
Hence to increase the level of GABA in the brain in order to prevent the
occurrence of seizure disorders, either one of the following steps should be adopted:
Spontaneous opening
spill
GAT-1
Downregulation GABAB
of tonic receptors receptor
Fig. 8.15 Various mechanisms altering the activity of tonic GABAA receptors. Activated by four
mechanisms: (1) spill of synaptically released GABA into extrasynaptic space due to insufficient
clearance by GATs, (2) GAT reversal causes nonvesicular release, (3) spontaneous opening in
absence of extracellular GABA, (4) GABAB receptor activation increases tonic GABAA signaling
via an intracellular mechanism
the disease which may fadeout after years of successful control (Upadhyay et al.
2017) as shown in Table 8.13.
Around 70 years ago, the discovery of neurotransmitter GABA was made and in this
long passage of time, several findings including agonists and antagonists have proven
it to be milestone in the scientific research field. Later, the identification of its
receptors GABAA and GABAB made the elucidation of targets and therapeutic
outcomes more significant. Today, the success of a number of molecules, specifi-
cally the clinical reliability of benzodiazepines and antiepileptic drugs, strongly
attests to the remarked contribution of neurotransmitter GABA in neuro research.
Initially, the benefits of GABA were assessed for only its calming, inhibitory, and
impaired motor-oriented function but now also get diverted towards its role in
neuron survival, memory, and other disease modifying functions. For example,
nowadays the role of GABA receptor is getting explored for altered synapse function
in AD. It is also expected to be a reliable biomarker for earlier diagnosis of disease in
8 Pharmacology of GABA and Its Receptors 283
dementia patients. Moreover, the usual side effects of BZDs also get eliminated as
in-depth structural elucidation of receptors takes place. The knowledge regarding the
different subunits of GABA and its functions raises more possibility with high
accuracy and fewer side effects. Recently, the work is ongoing upon the different
subunits of GABAA receptor for limiting side effects of anxiolytic drugs. Previously,
the target of anxiolytic drug was the interface of α and γ subunits but it shows
sedation rather than calmness. But now the specific agonists and partial agonists of
α2, α3, and α5 subunits are getting evaluated for therapeutic efficacy. Apart from
this, the role of these subunits is also getting explored for other therapeutic actions
including memory, neurotoxicity, and neuron survival in multiple neurodegenerative
disorders. The success of such findings and efforts could be analyzed from a plenty
of molecules undergoing evaluation for GABA modifying properties under recent
clinical trials (Schanzer et al. 2019; Zahn et al. 2019; Nikmaram et al. 2017) as
shown in Table 8.14.
284 S. Kaur et al.
8.9 Conclusion
GABA is one of the major key players of inhibitory neurotransmission in the central
nervous system and observed to be widely distributed throughout the mammalian
brain. The GABAA receptor influences have been studied for decades and are
beginning to be well understood; research on the GABAB and GABAC receptors
are more recent and currently growing. The crucial regulatory functions of GABAB
have become increasingly evident, whereas GABAC receptor studies are only at the
beginning stage. GABA acts through their receptors and helps to control
the excitability of cortical networks by modulating a network of cortical interneurons
and useful in various CNS-related disorders including Parkinson’s disease, epilepsy,
anxiety, schizophrenia, and dementia. Numerous anxiolytic, sedative, and hypnotics
show their molecular signaling through GABA receptors and dominantly emerge as
powerful drug targets. GABAC receptors show a lower threshold of activation than
GABAA and GABAB receptors, an essential property that is rich in promise for
future clinical applications. Hence, exploring the more molecular mechanisms may
result in a better understanding of their role in CNS disorder.
8 Pharmacology of GABA and Its Receptors 285
References
Abdel-Wahab BA, Metwally ME (2011) Ginkgo biloba enhances the anticonvulsant and
neuroprotective effects of sodium valproate against kainic acid-induced seizures in mice. J
Pharmacol Toxicol 6(8):679–690
Albus H et al (2005) A GABAergic mechanism is necessary for coupling dissociable ventral and
dorsal regional oscillators within the circadian clock. Curr Biol 15(10):886–893. Elsevier
Alexander GE (2004) Biology of Parkinson’s disease: pathogenesis and pathophysiology of a
multisystem neurodegenerative disorder. Dialogues Clin Neurosci 6(3):259. Les Laboratoires
Servier
Allen KL et al (2009) Cannabinoid (CB1), GABAA and GABAB receptor subunit changes in the
globus pallidus in Huntington’s disease. J Chem Neuroanat 37(4):266–281. Elsevier
Anaclet C, Fuller PM (2017) Brainstem regulation of slow-wave-sleep. Curr Opin Neurobiol
44:139–143. Elsevier
Andrews G et al (2010) Generalized worry disorder: a review of DSM-IV generalized anxiety
disorder and options for DSM-V. Depress anxiety 27(2):134–147. Wiley Online Library
Appel K et al (2011) Modulation of the γ-aminobutyric acid (GABA) system by Passiflora incarnata
L. Phytotherapy Res 25(6):838–843. Wiley Online Library
Arnaud C, Gauthier P, Gottesmann C (2001) Study of a GABA C receptor antagonist on sleep-
waking behavior in rats. Psychopharmacology 154(4):415–419. Springer
Aroniadou-Anderjaska V, Qashu F, Braga MFM (2007) Mechanisms regulating GABAergic
inhibitory transmission in the basolateral amygdala: implications for epilepsy and anxiety
disorders. Amino acids 32(3):305–315. Springer
Badawy RAB, Harvey AS, Macdonell RAL (2009) Cortical hyperexcitability and epileptogenesis:
understanding the mechanisms of epilepsy–part 1. J Clin Neurosci 16(3):355–365. Elsevier
Barber RP et al (1978) GABAergic terminals are presynaptic to primary afferent terminals in the
substantia gelatinosa of the rat spinal cord. Brain Res 141(1):35–55. Elsevier
Bateson AN (2004) The benzodiazepine site of the GABAA receptor: an old target with new
potential? Sleep Med 5:S9–S15. Elsevier
Ben-Ari Y (2002) Excitatory actions of gaba during development: the nature of the nurture. Nat Rev
Neurosci 3(9):728. Nature Publishing Group
Bentzen BH, Grunnet M (2011) Central and peripheral GABAA receptor regulation of the heart rate
depends on the conscious state of the animal. Adv Pharmacol Sciences 2011. https://doi.org/10.
1155/2011/578273. Hindawi
Besag FMC, Patsalos PN (2012) New developments in the treatment of partial-onset epilepsy.
Neuropsychiatr Dis Treat 8:455. Dove Press
Bettler B et al (2004) Molecular structure and physiological functions of GABAB receptors,
Physiological reviews. Am Physiol Soc 84(3):835–867
Billinton A et al (2000) GABAB receptor heterodimer-component localisation in human brain. Mol
Brain Res 77(1):111–124. Elsevier
Birnir B, Korpi ER (2007) The impact of sub-cellular location and intracellular neuronal proteins on
properties of GABAA receptors. Curr Pharm Design 13(31):3169–3177. Bentham Science
Publishers
Blackburn T (2010) Gabab receptor pharmacology: a tribute to Norman Bowery. Academic Press,
Cambridge
Błaszczyk JW (2016) Parkinson’s disease and neurodegeneration: GABA-collapse hypothesis.
Front Neurosci 10:269. Frontiers
Bormann J (2000) The ‘ABC’ of GABA receptors. Trends Pharmacol Sci 21(1):16–19. Elsevier
Bowery NG, Smart TG (2006) GABA and glycine as neurotransmitters: a brief history. Br J
Pharmacol 147(S1):S109–S119. Wiley Online Library
Bowery NG, Hill DR, Hudson AL (1982) Guanyl nucleotides decrease the binding affinity of
GABAB but not GABAA receptors in the mammalian CNS. Br J Pharmac 75(Suppl. 86P)
Browne TR, Holmes GL (2008) Handbook of epilepsy. Jones & Bartlett Learning, Burlington, MA
286 S. Kaur et al.
Byrne LM, Wild EJ (2016) Cerebrospinal fluid biomarkers for Huntington’s disease. J Huntington’s
Disease 5(1):1–13. IOS Press
Caldji C et al (2004) Maternal behavior regulates benzodiazepine/GABA A receptor subunit
expression in brain regions associated with fear in BALB/c and C57BL/6 mice.
Neuropsychopharmacology 29(7):1344. Nature Publishing Group
Carver CM, Reddy DS (2013) Neurosteroid interactions with synaptic and extrasynaptic GABA A
receptors: regulation of subunit plasticity, phasic and tonic inhibition, and neuronal network
excitability. Psychopharmacology 230(2):151–188. Springer
Castelli MP et al (2012) Characterization of COR627 and COR628, two novel positive allosteric
modulators of the GABAB receptor. J Pharmacol Exp Ther 340(3):529–538. ASPET
Cawley N et al (2015) Reduced gamma-aminobutyric acid concentration is associated with physical
disability in progressive multiple sclerosis. Brain 138(9):2584–2595. Oxford University Press
Chambers CD, Stokes MG, Mattingley JB (2004) Modality-specific control of strategic spatial
attention in parietal cortex. Neuron 44(6):925–930. Elsevier
Chapouthier G, Venault P (2002) GABA-A receptor complex and memory processes. Curr Top
Med Chem 2(8):841–851. Bentham Science Publishers
Charych EI et al (2009) GABAA receptors and their associated proteins: implications in the
etiology and treatment of schizophrenia and related disorders. Neuropharmacology 57
(5–6):481–495. Elsevier
Chebib M, Johnston GAR (2000) GABA-activated ligand gated ion channels: medicinal chemistry
and molecular biology. J Med Chem 43(8):1427–1447. ACS Publications
Chebib M et al (2009) Novel, potent, and selective GABAC antagonists inhibit myopia develop-
ment and facilitate learning and memory. J Pharmacol Exp Ther 328(2):448–457. ASPET
Chen Y et al (2007) Study on olfactory function in GABAC receptor/channel ρ1 subunit knockout
mice. Neurosci Lett 427(1):10–15. Elsevier
Chuhma N et al (2009) Dopamine neuron glutamate cotransmission: frequency-dependent modula-
tion in the mesoventromedial projection. Neuroscie 164(3):1068–1083. Elsevier
Ciranna áL (2006) Serotonin as a modulator of glutamate-and GABA-mediated neurotransmission:
implications in physiological functions and in pathology. Curr Neuropharmacol 4(2):101–114.
Bentham Science Publishers
Collares EF, Vinagre AM (2005) Effect of the GABA B agonist baclofen on dipyrone-induced
delayed gastric emptying in rats. Braz J Med Biol Res 38(1):99–104. SciELO Brasil
Cooper JR, Bloom FE, Roth RH (2003) The biochemical basis of neuropharmacology. Oxford
University Press, USA
Crestani F et al (2000) Mechanism of action of the hypnotic zolpidem in vivo. Br J Pharmacol 131
(7):1251–1254. Wiley Online Library
Cryan JF et al (2004) Behavioral characterization of the novel GABAB receptor-positive modulator
GS39783 (N, N0 -dicyclopentyl-2-methylsulfanyl-5-nitro-pyrimidine-4, 6-diamine): anxiolytic-
like activity without side effects associated with baclofen or benzodiazepines. J Pharmacol Exp
Ther 310(3):952–963. ASPET
Cutting GR et al (1992) Identification of a putative γ-aminobutyric acid (GABA) receptor subunit
rho2 cDNA and colocalization of the genes encoding rho2 (GABRR2) and rho1 (GABRR1) to
human chromosome 6q14–q21 and mouse chromosome 4. Genomics 12(4):801–806. Elsevier
Derwenskus J (2011) Current disease-modifying treatment of multiple sclerosis. Mt Sinai J Med 78
(2):161–175. Wiley Online Library
Dézsi L, Vécsei L (2011) Parkinson’s disease: will therapy move beyond dopaminergic medica-
tion? Clin Invest 1(3):381–398
Drasbek KR, Christensen J, Jensen K (2006) Gamma-hydroxybutyrate–a drug of abuse. Acta
Neurol Scand 114(3):145–156. Wiley Online Library
Du J-L, Yang X-L (2000) Subcellular localization and complements of GABAA and GABAC
receptors on bullfrog retinal bipolar cells. J Neurophysiol 84(2):666–676. American Physiolog-
ical Society Bethesda, MD
8 Pharmacology of GABA and Its Receptors 287
Kantamneni S et al (2008) GISP binding to TSG101 increases GABAB receptor stability by down-
regulating ESCRT-mediated lysosomal degradation. J Neurochem 107(1):86–95. Wiley Online
Library
Kaupmann K et al (1997) Expression cloning of GABA B receptors uncovers similarity to
metabotropic glutamate receptors. Nature 386(6622):239–246. Nature Publishing Group
Keberle H, Faigle JW, Wilhelm M (1968) Procedure for the preparation of new amino acids, Swiss
Patent 449046, Publication Date: 11 April 1968, Priority: 9 July 1963, In Chem Abstr
Kerr DIB, Ong J (1996) GABAB receptors: targets for drug development. Drug Discov Today 1
(9):371–380. Elsevier
Kesby JP et al (2018) Dopamine, psychosis and schizophrenia: the widening gap between basic and
clinical neuroscience. Transl Psychiatry 8(1):30. Nature Publishing Group
Khazipov R (2016) GABAergic synchronization in epilepsy. Cold Spring Harb Perspect Med 6(2):
a022764. Cold Spring Harbor Laboratory Press
Kilb W (2012) Development of the GABAergic system from birth to adolescence. Neuroscientist 18
(6):613–630. SAGE Publications Sage CA: Los Angeles, CA
Kim DH et al (2012) GABA a receptor blockade enhances memory consolidation by increasing
hippocampal BDNF levels. Neuropsychopharmacology 37(2):422. Nature Publishing Group
Kochanek PM et al (2015) Emerging therapies in traumatic brain injury. In: Seminars in neurology.
Thieme Medical Publishers, New York, NY, pp 83–100
Korol SV et al (2015) GLP-1 and exendin-4 transiently enhance GABAA receptor–mediated
synaptic and tonic currents in rat hippocampal CA3 pyramidal neurons. Diabetes 64
(1):79–89. Am Diabetes Assoc
Kuffler SW (1954) Mechanisms of activation and motor control of stretch receptors in lobster and
crayfish. J Neurophysiol 17(6):558–574
Kumar A, Singh A (2015) A review on Alzheimer’s disease pathophysiology and its management:
an update. Pharmacol Rep 67(2):195–203. Elsevier
Laruelle M (2014) Schizophrenia: from dopaminergic to glutamatergic interventions. Curr Opin
Pharmacol 14:97–102. Elsevier
Lasoń W, Chlebicka M, Rejdak K (2013) Research advances in basic mechanisms of seizures and
antiepileptic drug action. Pharmacol Rep 65(4):787–801. Elsevier
Lazzari C et al (2015) Aβ42 oligomers selectively disrupt neuronal calcium release. Neurobiol
Aging 36(2):877–885. Elsevier
Lee S, Kim K, Zhou ZJ (2010) Role of ACh-GABA cotransmission in detecting image motion and
motion direction. Neuron 68(6):1159–1172. Elsevier
Lewis DA et al (2012) Cortical parvalbumin interneurons and cognitive dysfunction in schizophre-
nia. Trends Neurosci 35(1):57–67. Elsevier
Li J-T et al (2016) Repeated blockade of NMDA receptors during adolescence impairs reversal
learning and disrupts GABAergic interneurons in rat medial prefrontal cortex. Front Mol
Neurosci 9:17. Frontiers
Lie MEK et al (2017) Glial GABA transporters as modulators of inhibitory signalling in epilepsy
and stroke. In: Glial amino acid transporters. Springer, New York, NY, pp 137–167
Lin E et al (2014) Optical mapping of the electrical activity of isolated adult zebrafish hearts: acute
effects of temperature. Am J Physiol Regul Integr Comp Physiol 306(11):R823–R836. Ameri-
can Physiological Society Bethesda, MD
Loma I, Heyman R (2011) Multiple sclerosis: pathogenesis and treatment. Curr Neuropharmacol 9
(3):409–416. Bentham Science Publishers
López-Chávez A, Miledi R, Martínez-Torres A (2005) Cloning and functional expression of the
bovine GABAC ρ2 subunit: molecular evidence of a widespread distribution in the CNS.
Neurosci Res 53(4):421–427. Elsevier
Löscher W et al (2013) New avenues for anti-epileptic drug discovery and development. Nat Rev
Drug Discov 12(10):757–776. Nature Publishing Group
Lukasiewicz PD et al (2004) GABAC receptor-mediated inhibition in the retina. Vision Res 44
(28):3289–3296. Elsevier
8 Pharmacology of GABA and Its Receptors 289
Ma P et al (2015) Effect of GABA on blood pressure and blood dynamics of anesthetic rats. Int J
Clin Exp Med 8(8):14296. e-Century Publishing Corporation
Ma C et al (2019) Sleep regulation by neurotensinergic neurons in a thalamo-amygdala circuit.
Neuron. https://doi.org/10.1016/j.neuron.2019.05.015. Elsevier
Macey PM et al (2016) Obstructive sleep apnea is associated with low GABA and high glutamate in
the insular cortex. J Sleep Res 25(4):390–394. Wiley Online Library
Madikians A, Giza CC (2006) A clinician’s guide to the pathophysiology of traumatic brain injury.
Indian J Neurotrauma 3(01):9–17. Thieme Medical and Scientific Publishers Private Ltd.
Maffucci JA, Gore AC (2009) Hypothalamic neural systems controlling the female reproductive life
cycle: gonadotropin-releasing hormone, glutamate, and GABA. Int Rev Cell Mol Biol
274:69–127. Elsevier
Maiti P, Gregg LC, McDonald MP (2016) MPTP-induced executive dysfunction is associated with
altered prefrontal serotonergic function. Behav Brain Res 298:192–201. Elsevier
Makkar SR, Zhang SQ, Cranney J (2012) Behavioral and neural analysis of GABA in the
acquisition, consolidation, reconsolidation, and extinction of fear memory: Corrigendum.
Neuropsychopharmacology 35(8):1625–1652. Nature Publishing Group
Mandolesi G et al (2015) Synaptopathy connects inflammation and neurodegeneration in multiple
sclerosis. Nat Rev Neurol 11(12):711. Nature Publishing Group
Martin DL, Olsen RW (2000) GABA in the nervous system: the view at fifty years. Lippincott
Williams and Wilkins
Martin SC, Russek SJ, Farb DH (2001) Human GABABR genomic structure: evidence for splice
variants in GABABR1 but not GABABR2. Gene 278(1–2):63–79. Elsevier
Masiulis S et al (2019) GABA A receptor signalling mechanisms revealed by structural pharma-
cology. Nature 565(7740):454–459. Nature Publishing Group
Mason SL, Barker RA (2009) Emerging drug therapies in Huntington’s disease. Expert Opin Emerg
Drugs 14(2):273–297. Taylor & Francis
McDonnell MN, Orekhov Y, Ziemann U (2007) Suppression of LTP-like plasticity in human motor
cortex by the GABA B receptor agonist baclofen. Exp Brain Res 180(1):181–186. Springer
Mingote S et al (2017) Dopamine neuron dependent behaviors mediated by glutamate
cotransmission. Elife 6:e27566. eLife Sciences Publications Limited
Möhler H (2012) The GABA system in anxiety and depression and its therapeutic potential.
Neuropharmacology 62(1):42–53. Elsevier
Morales B, Choi S-Y, Kirkwood A (2002) Dark rearing alters the development of GABAergic
transmission in visual cortex. J Neurosci 22(18):8084–8090. Soc Neuroscience
Naffaa MM et al (2017) GABA-ρ receptors: distinctive functions and molecular pharmacology. Br J
Pharmacol 174(13):1881–1894. Wiley Online Library
Nakazawa K et al (2012) GABAergic interneuron origin of schizophrenia pathophysiology.
Neuropharmacology 62(3):1574–1583. Elsevier
Nguyen R et al (2014) Parvalbumin and GAD65 interneuron inhibition in the ventral hippocampus
induces distinct behavioral deficits relevant to schizophrenia. J Neurosci 34(45):14948–14960.
Soc Neuroscience
Nikmaram N et al (2017) Recent advances in γ-aminobutyric acid (GABA) properties in pulses: an
overview. J Sci Food Agric 97(9):2681–2689. Wiley Online Library
Nowak G et al (2006) Antidepressant-like activity of CGP 36742 and CGP 51176, selective
GABAB receptor antagonists, in rodents. Br J Pharmacol 149(5):581–590. Wiley Online
Library
Nuss P (2015) Anxiety disorders and GABA neurotransmission: a disturbance of modulation.
Neuropsychiatr Dis Treat 11:165. Dove Press
Olsen RW, Sieghart W (2009) GABAA receptors: subtypes provide diversity of function and
pharmacology. Neuropharmacology 56(1):141–148. Elsevier
Orts-Del’Immagine A, Pugh JR (2018) Activity-dependent plasticity of presynaptic GABAB
receptors at parallel fiber synapses. Synapse 72(5):e22027. Wiley Online Library
290 S. Kaur et al.
Pin J-P, Prézeau L (2007) Allosteric modulators of GABAB receptors: mechanism of action and
therapeutic perspective. Curr Neuropharmacol 5(3):195–201. Bentham Science Publishers
Pinard A, Seddik R, Bettler B (2010) GABAB receptors: physiological functions and mechanisms
of diversity. In: Advances in pharmacology. Elsevier, Amsterdam, pp 231–255
Plante DT, Jensen JE Winkelman JW (2012) The role of GABA in primary insomnia. Sleep 35
(6):741–742. Oxford University Press
Polenzani L, Woodward RM, Miledi R (1991) Expression of mammalian gamma-aminobutyric
acid receptors with distinct pharmacology in Xenopus oocytes. Proc Natl Acad Sci 88
(10):4318–4322. Natl Acad Sci
Prud’homme G, Glinka Y, Wang Q (2013) GABA exerts anti-inflammatory and immunosuppres-
sive effects (P5175). Am Assoc Immnol
Rana N et al (2010) Caffeine-induced effects on heart rate in zebrafish embryos and possible
mechanisms of action: an effective system for experiments in chemical biology. Zebrafish 7
(1):69–81. Mary Ann Liebert, Inc. 140 Huguenot Street, 3rd Floor New Rochelle, NY 10801
USA
Rao T, Zhang C (2011) 3, 4-methylenedioxyphenyl inhibitors of gaba aminotransferase and/or gaba
reuptake transporter inhibitor. Google Patents
Riordan AJ et al (2017) Estradiol and luteinizing hormone reverse memory loss in phencyclidine
model of schizophrenia: evidence for hippocampal GABA action. bioRxiv, p 207159. Cold
Spring Harbor Laboratory
Roberts E, Frankel S (1950) γ-aminobutyric acid in brain: its formation from glutamic acid. J Biol
Chem 187:55–63
Rosenberg M (2012) Diagnosis, treatment options, and costs of schizophrenia. J Manage Care Med
3:10–15
Roth FC, Draguhn A (2012) GABA metabolism and transport: effects on synaptic efficacy. Neural
Plast 2012:805830. Hindawi
Rowley NM et al (2012) Glutamate and GABA synthesis, release, transport and metabolism as
targets for seizure control. Neurochem Int 61(4):546–558. Elsevier
Rubinsztein DC (2006) The roles of intracellular protein-degradation pathways in
neurodegeneration. Nature 443(7113):780. Nature Publishing Group
Rudolph U, Möhler H (2014) GABAA receptor subtypes: therapeutic potential in Down syndrome,
affective disorders, schizophrenia, and autism. Annu Rev Pharmacol Toxicol 54:483–507.
Annual Reviews
Rudolph U et al (2000) erratum: Benzodiazepine actions mediated by specific γ-aminobutyric acid
A receptor subtypes. Nature 404(6778):629. Nature Publishing Group
Saito S et al (2010) Interaction between hippocampal γ-aminobutyric acidA and N-methyl-D-
aspartate receptors in the retention of spatial working memory in rats. Biol Pharm Bull 33
(3):439–443. The Pharmaceutical Society of Japan
Sałat K, Kulig K (2011) GABA transporters as targets for new drugs. Future Med Chem 3
(2):211–222. Future Science
Schanzer B et al (2019) Novel investigational therapeutics for generalized anxiety disorder (GAD).
Expert Opin Investig Drugs. https://doi.org/10.1080/13543784.2019.1680638. Taylor &
Francis
Schipper S et al (2016) Tonic GABA A receptors as potential target for the treatment of temporal
lobe epilepsy. Mol Neurobiol 53(8):5252–5265. Springer
Schofield PR et al (1987) Sequence and functional expression of the GABA A receptor shows a
ligand-gated receptor super-family. Nature 328(6127):221–227. Nature Publishing Group
Scotter EL, Abood ME, Glass M (2010) The endocannabinoid system as a target for the treatment of
neurodegenerative disease. Br J Pharmacol 160(3):480–498. Wiley Online Library
Selten MM et al (2016) Increased GABA B receptor signaling in a rat model for schizophrenia. Sci
Rep 6:34240. Nature Publishing Group
8 Pharmacology of GABA and Its Receptors 291
Semyanov AV (2002) GABA-ergic inhibition in the CNS: types of GABA receptors and
mechanisms of tonic GABA-mediated inhibitory action. Neurophysiology 34(1):71–80.
Springer
Shaban H et al (2006) Generalization of amygdala LTP and conditioned fear in the absence of
presynaptic inhibition. Nat Neurosci 9(8):1028. Nature Publishing Group
Sharpley CF (2009) Malfunction in GABA and glutamate as pathways to depression: a review of
the evidence. Clin Med 1, p. CMT-S3481. Therapeutics. SAGE Publications Sage UK: London,
England
Shin LM, Liberzon I (2010) The neurocircuitry of fear, stress, and anxiety disorders.
Neuropsychopharmacology 35(1):169. Nature Publishing Group
Shingai R et al (1997) Functional expression of GABA rho 3 receptors in Xenopus oocytes.
Neurosci Res 26:387–390
Sieghart W et al (2012) A novel GABAA receptor pharmacology: drugs interacting with the α+
β-interface. Br J Pharmacol 166(2):476–485. Wiley Online Library
Sigel E, Steinmann ME (2012) Structure, function, and modulation of GABAA receptors. J Biol
Chem 287(48):40224–40231. ASBMB
Silverman RB (2008) From basic science to blockbuster drug: the discovery of Lyrica. Angew
Chem Int Ed 47(19):3500–3504. Wiley Online Library
Silverman RB (2012) The 2011 EB Hershberg Award for important discoveries in medicinally
active substances:(1 S, 3 S)-3-amino-4-difluoromethylenyl-1-cyclopentanoic acid (CPP-115), a
GABA aminotransferase inactivator and new treatment for drug addiction and infantile spasms.
J Med Chem 55(2):567–575. ACS Publications
Solas M, Puerta E, J Ramirez M (2015) Treatment options in Alzheimer s disease: the GABA story.
Curr Pharm Design 21(34):4960–4971. Bentham Science Publishers
Steenbergen L et al (2015) γ-Aminobutyric acid (GABA) administration improves action selection
processes: a randomised controlled trial. Sci Rep 5(1):1–7. Nature Publishing Group
Strandwitz P et al (2019) GABA-modulating bacteria of the human gut microbiota. Nat Microbiol
4(3):396–403. Nature Publishing Group
Tadavarty R et al (2015) Are presynaptic GABA-Cρ2 receptors involved in anti-nociception?
Neurosci Lett 606:145–150. Elsevier
Tallman JF, Smith CC, Henneberry RC (1977) Induction of functional beta-adrenergic receptors in
HeLa cells. Proc Natl Acad Sci 74(3):873–877. Natl Acad Sci
Treiman DM (2001) GABAergic mechanisms in epilepsy. Epilepsia 42:8–12. Wiley Online Library
Upadhyay J, Upadhyay G, Rana AJ (2017) A prospective study on prevalence of epilepsy disorders
and drug utilization pattern. Asian J Pharm Clin Res 10(3):136–139
Uusi-Oukari M, Korpi ER (2010) Regulation of GABAA receptor subunit expression by pharma-
cological agents. Pharmacol Rev 62(1):97–135. ASPET
van Vliet EA et al (2018) Neuroinflammatory pathways as treatment targets and biomarker
candidates in epilepsy: emerging evidence from preclinical and clinical studies. Neuropathol
Appl Neurobiol 44(1):91–111. Wiley Online Library
Vinkers CH et al (2010) The inhibitory GABA system as a therapeutic target for cognitive
symptoms in schizophrenia: investigational agents in the pipeline. Expert Opin Investig Drugs
19(10):1217–1233. Taylor & Francis
Waldvogel HJ, Faull RLM (2015) The diversity of GABAA receptor subunit distribution in the
normal and Huntington’s disease human Brain1. In: Advances in pharmacology. Elsevier,
Amsterdam, pp 223–264
Wu C, Sun D (2015) GABA receptors in brain development, function, and injury. Metabolic brain
disease 30(2):367–379. Springer
Wu Z et al (2014) Tonic inhibition in dentate gyrus impairs long-term potentiation and memory in
an Alzheimer’s disease model. Nat Commun 5(1):1–13. Nature Publishing Group
Yadav RK, Khanday MA, Mallick BN (2019) Interplay of dopamine and GABA in substantia nigra
for the regulation of rapid eye movement sleep in rats. Behav Brain Res 376:112169. Springer
292 S. Kaur et al.
Yi J-H, Hazell AS (2006) Excitotoxic mechanisms and the role of astrocytic glutamate transporters
in traumatic brain injury. Neurochem Int 48(5):394–403. Springer
Zahn NM et al (2019) MIDD 0301–A first-in-class anti-inflammatory asthma drug targets GABAA
receptors without causing systemic immune suppression. Basic Clin Pharmacol Toxicol. https://
doi.org/10.1111/bcpt.13206. Wiley Online Library
Zhang D et al (2001) Structure and function of GABAC receptors: a comparison of native versus
recombinant receptors. Trends Pharmacol Sci 22(3):121–132. Elsevier
Zhang C, Liu X, Yuan TF (2017) Somatostatin-positive GABAergic interneuron: new targets for
depression. Nature Publishing Group
Pharmacology of Melatonin and Its
Receptors 9
Shamsher Singh, Arti Rana, Sunpreet Kaur, Jasdeep Singh,
Vikrant Rahi, Hira Choudhury, and Puneet Kumar
Abstract
Keywords
Abbreviation
4P-ADOT 4-Phenyl-2-acetamidotetralin
4P-PDOT 4-Phenyl-2-acetamidotetralin
5-HT 5-Hydroxytryptamine
5-MCA-NAT 5-Methoxycarbonylamino-N-acetyltryptamine
6-SMT 6-Sulphatoxymelatonin
Ab Amyloid β
AD Alzheimer’s disease
AFMK N-acetyl-N-formyl-5-methoxy kynurenine
AMPA α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptor
ATP Adenosine triphosphate
Bcl-2 β-cell lymphoma 2
BDNF Brain-derived neurotrophic factor
BKCa Blocking of calcium-activated potassium channels
cAMP Cyclic adenosine monophosphate
CCK-2 Cholecystokinin-2
cGMP Cyclic guanosine monophosphate
CK1 Casein kinase 1
CK2 Casein kinase
CNS Central nervous system
Cox-2 Cyclooxygenase-2
CREB cAMP response element-binding protein
CVS Melatonin actions on the cardiovascular system
DNA Deoxyribonucleic acid
ERK Extracellular signal-regulated kinase
ETC Electron transport chain
FSH Follicle-stimulating hormone
GABA Gamma-aminobutyric acid
GDNF Glial cell-derived neurotrophic factor
GDNF Glial cell line-derived neurotrophic factor
GIT Gastrointestinal tract
GPCR G-protein-coupled receptor
GPx Glutathione peroxidase
GRd Glutathione reductase
9 Pharmacology of Melatonin and Its Receptors 295
9.1 Introduction
9.1.1 History
Before the discovery of melatonin, researchers used the injection of pineal gland
extract to find out the role of pineal gland in different biological processes of the
body. In some studies the researchers used pineal gland extract in tadpoles, frogs,
toads, and fish to reveal the skin lightening effect of pineal gland biochemicals
(Whitlock et al. 1954). In 1958, Aaron B. Lerner revealed the role of a neurochemi-
cal which reverses the darkening effect of the melanocyte stimulating hormone and
named it “Melatonin” (Lerner et al. 1958) (Table 9.1).
Table 9.1 Historical development in the discovery of melatonin and its receptors
Name Year Discoveries Reference
Herophilos 325–280 First time discovering the pineal Kappers
BC organ in man (1979)
Aaron Lerner 1958 Discovered the chemical structure Lerner et al.
of melatonin (1958)
Bubenik 1993 Found melatonin in human Filadelfi and
intestine and the retina of many Castrucci
nonmammalian vertebrates (1996)
Miles, Stankov and Reiter, 1989; The highly specific ligand 2 [‘25I]- Filadelfi and
Stankov Kennaway and 1990; iodomelatonin allowed the Castrucci
Hugel, Almeida 1991; expansion of receptor investigation (1996)
1992; 1951 to peripheral tissues, such as retina,
spleen, gastrointestinal tract,
uterus, ovary, liver, and cultured
normal and tumor cells
Dubocovich 1998, 1981 Melatonin receptors were first Dubocovich
1988, 1995 classified according to classic et al. (2010)
1990,1994 pharmacological criteria
Nosjean 2000 Identification and characterization Nosjean
of a third membrane-bound et al. (2000)
melatonin binding site MT3
receptor
Ebisawa 1994 The MT1 receptor (Mel1c) was Ebisawa
first cloned in frogs et al. (1994)
Reppert 1995 The MT2 receptor was cloned from Reppert
the brain, retina, and human et al. (1995)
pituitary gland
Ayoub 2004 Both MT1 and MT2 can form Ayoub et al.
homo and heterodimers (2004)
Takeda Pharmaceutical 2005 Development of melatonin Neubauer
Company receptor agonist TAK-375, now (2008)
known as ramelteon
Christian de Bodinat 2010 Agomelatine, the first melatonergic De Bodinat
antidepressant et al. (2010)
Daniel P. Cardinali 2013 Role of melatonin and its analogs Cardinali
in insomnia and depression et al. (2012)
Gabriella Gobbi 2019 Differential function of melatonin Gobbi and
MT1 and MT2 receptors in REM Comai
and NREM sleep (2019)
level of pineal melatonin is being observed late in the evening which reaches
maximum up to 4:00 a.m. (2:00 and 4:00 a.m. is a maximum observed range) and
then gets back down to the normal daytime levels. It is very difficult to detect the
daytime level of melatonin. In contrast to sunlight, artificial lights are sufficient to
block the nocturnal release of melatonin (Peuhkuri et al. 2012).
Melatonin possesses rapid metabolism with a half-life that varies between 10 and
60 min in humans following exogenous administration. It is catabolized mostly by
the liver and gets excreted in the urine. Metabolite like 6-sulphatoxymelatonin
9 Pharmacology of Melatonin and Its Receptors 299
L-Tryptophan
Tryptophan
5-hydroxylase
5-hydroxytryptophan
Aromatic L-amino
acid decarboxylase
Serotonin
Serotonin N-
acetyltransferase
N- acetyl serotonin
Hydroxyindole-O-methyl
transferase (HIOMT)
Melatonin
(6-SMT) in urine indicates that melatonin status can be evaluated melatonin in the
overnight urine sample (Peuhkuri et al. 2012).
Two distinct classes of melatonin receptors are found in different regions of the body,
which are expressed in humans and have been reported so far, MT1 and MT2,
formerly designated as Mel1a and Mel1b, respectively (Dubocovich et al. 2003).
The majority of the melatonin receptors are found in the following regions of the
300 S. Singh et al.
MT1 receptors are high-affinity receptors that fall into the GPCR superfamily and
the binding of melatonin to these receptors inhibits the activity of adenylate cyclase
activity in target cells (Pandi-Perumal et al. 2008). The two subgroups of the MT1
receptors are MT1a receptors and MT1b receptors. Cardiac vessels constriction and
circadian rhythms are modulated by MT1 melatonin receptor generally found in
9 Pharmacology of Melatonin and Its Receptors 301
Table 9.2 Functions of melatonin receptors based on location in the different regions of the body
Receptor
Location type Function Reference
CNS
Hippocampus MT1, MT2 Inhibition of neuronal activity and Savaskan et al. (2005,
excitatory responses in memory 2002)
Modification in Alzheimer disease
Enhancement in seizure threshold via
suppression in GABAA-receptor
function
Cerebellum MT1, MT2 Involvement and interaction with Al-Ghoul et al. (1998)
glutamatergic synapse
Various MT1, MT2 Dopamine release evoked by Reppert et al. (1995)
retinal cell stimulation gets inhibited
Adaptation to low light intensity
Modification in photoreceptor
functions rod phototransduction
pathways
SCN MT1 Initiation and improvisation in sleep
Modification of circadian rhythm
(blind people, phase shift worker, jet
lag)
Central MT1 Modulation of dopamine synthesis and Uz et al. (2005)
dopaminergic release
system Increased sensitivity and activation of
dopamine receptors
CVS
Cerebral MT1 Unknown effects Savaskan et al. (2001)
arteries
Aorta MT1, MT2 Vasodilation may occur Ekmekcioglu et al.
(2003)
Cardiac MT1, MT2 Modulation of beta-adrenergic Ekmekcioglu et al.
ventricular receptor-mediated cAMP signaling (2003, 2001)
wall processes may be initiated
Increased stimulation of voltage-
activated calcium current
Negative inotropic effects may occur
Coronary MT1, MT2 MT2 receptors-mediated vasodilation Ekmekcioglu (2006);
arteries MT1 receptors-mediated Ekmekcioglu et al.
vasoconstriction (2003, 2001)
GIT
Duodenal MT2 Stimulation of HCO3–secretion via Sjöblom et al. (2001);
enterocytes neural stimulation Sjöblom and
Flemström (2003)
Pancreatic MT1 Acid/base homeostasis regulation Ekmekcioglu (2006)
cancer cell (stimulation of HCO3–secretion)
lines
Gallbladder MT1 Gallbladder contraction may occur Aust et al. (2004)
epithelia
302 S. Singh et al.
SCN and cardiac vessels. Not only in these regions but these sets of receptors are
also found in peripheral tissues as well as other parts of the brain.
The MT2 receptors are low-affinity receptors that are coupled to phosphoinositol
hydrolysis. MT2 receptors are involved in retinal physiology, modulating circadian
rhythms, dilating cardiac vessels, and affecting inflammatory responses in the
microcirculation (Pandi-Perumal et al. 2008). Unlike the MT1, the restriction
is higher with regard to their localization; the involving regions are SCN of the
hypothalamus, cerebellum, cardiac vessels, kidney, ovary, retina, and other multiple
cell lines. The protein reflecting similar binding affinities to MT2 receptor is now
denoted as MT3 and is affinity purified from the Syrian hamster kidney (Nosjean
et al. 2000). It is found that an enzyme involved in detoxification named quinone
reductase 2 shares 95% homology to this enzyme. Lowering of intraocular pressure
and leukocyte adhesion induced by leukotriene B4 is inhibited by MT3 receptors
activation.
There are only one nuclear receptor and three membrane receptors.
Melatonin can scavenge nitrogen and oxygen-based reactants directly and along
with reducing the oxidative stress by multiple pathways. The relative importance of
the direct and indirect antioxidative processes of melatonin in vivo remains
unknown.
Two GPCRs mediate the physiological actions of melatonin, MT1 and MT2. In
sleep regulation, the role of MT1 and MT2 with high affinity in SCN is observed.
The neuronal firing rate is suppressed by MT1 receptor activation in the SCN, and on
the other hand circadian rhythm phase shifts are induced by MT2 receptors. In
peripheral organs both MT1 and MT2 receptors are present and make their contri-
bution to manage physiological functions. MT1 receptors activation pursue the
following events which include vasoconstriction of cerebral and peripheral arteries,
regulating the expression of Per1 gene in the anterior pituitary, and inhibiting the
secretion of prolactin from the pars tuberalis (Doghramji 2007). Various studies
support the involvement of MT2 receptors in the inhibition of dopamine release in
the retina, vasodilation, in retinal physiology, and also increasing splenocyte prolif-
eration. MT3 binding sites are widely distributed in the brain, liver, heart, kidneys,
and lungs. Several studies performed in vivo suggest its possible role in the regula-
tion of intraocular pressure and inflammatory responses inside the microvasculature.
9.5 Pharmacology
Several compounds have revealed the binding efficacy towards melatonin receptors
for significant therapeutic actions. Any compound having 100-fold or more binding
potential for specific receptor is called a ligand. Both the endogenous and exogenous
308 S. Singh et al.
melatonin proved to be potent agonists for MT1 and MT2 receptors providing
a variety of physiological responses (Dawoodi et al. 2012). Another compound
named as N-butanoyl-2-(2-methoxy-6H-isoindolo [2, 1-a] indol-11-yl)-ethanamine
(IIK7) that emerged as a selective MT2 melatonin receptor agonist proved to have
higher affinity towards MT2 in comparison to MT1. On the contrary, luzindole acts
as a selective antagonist for MT2 receptor as it shows a higher affinity towards MT2
in comparison to MT1 receptor. It provides significant results in circadian rhythm
disorders and depression-associated studies. Besides this, other compounds such as
4-phenyl-2-propionamidotetralin (4P-PDOT) and 4-phenyl-2- acetamidotetralin
(4P-ADOT) have also revealed significant antagonist properties for MT2 receptor
(Cecon et al. 2018). Research studies have shown that the success of selective
ligands for MT1 is less as compared to MT2 receptors as their efficacy and binding
affinity are low. To tackle this, a dimer of agomelatine is formed as ligand S26131
which shows 200-fold higher affinities for MT1 receptor in cell line studies (Liu
et al. 2016). Identical to these compounds, a number of different ligands have been
recognized for their specificity towards each melatonin receptor which is enlisted in
Table 9.3.
Moreover, the formation of MT1 or MT2 receptors with 5-HT2C provides suc-
cessful result in higher efficacy in various disorders. Such drugs include agomelatine
and TIK-301 which provide reliable results in depression studies (Liu et al. 2016).
Further on the basis of research data available, several melatonin targeting drugs are
successfully running for their therapeutic efficacy in different clinical conditions
(Dawoodi et al. 2012). Such compounds include ramelteon, agomelatine, TIK-301,
and tasimelteon that have gained clinical reliability for their efficacy in circadian
rhythms-associated disorders (Table 9.4).
Melatonin plays a very important role in ensuring that the organism is adjusted to
seasonal and environmental changes. Endogenous and exogenous melatonin play a
very important role in the control and coordination process. The release of endoge-
nous melatonin takes place in seasonal and circadian fashion. On the other hand,
behavioral and physiological responses are regulated by exogenous melatonin. In
this segment multiple mechanisms through which melatonin controls and
coordinates functions are to be discussed such as cardiovascular responses, endo-
crine functions, circadian rhythms, and immune system.
been explored for its in-depth analysis. The thalamic reticular nucleus is the major
region to coordinate sleep. Here, the localized MT1 receptor serves to modulate the
REM sleep whereas the localized MT2 receptor takes part in NREM sleep (Ng et al.
2017). The presence and activation of MT1 receptor in the retina regulates the
neurotransmitter release. Once melatonin get secreted from photoreceptors, it imme-
diately binds to melatonin receptors on amacrine cells and affects the subsequent
release of dopamine and GABA. Melatonin and dopamine act as a mutual
antagonists of one another. So, if melatonin binds on dopaminergic amacrine cells
then the subsequent release of dopamine decreases but if itself dopamine interacts
with D2 receptor then it leads to inhibition of melatonin release (Ng et al. 2017).
Normally, the binding of melatonin to MT1 receptor directly inhibits dopamine
release to facilitate dark adaptation.
the aircraft, dehydration, etc. (Arendt and Deacon 1997). Therefore, melatonin is
again considered as a good approach to treat time zone travelers; it improvises sleep
pattern without hindering daily performances and tasks.
Other effects: Other actions like the antidepressant action of melatonin is
confirmed after the discovery of agomelatine which acts synergistically via the
melatonin receptors and 5HT-2C receptors (Kasper and Hamon 2009). The drug
agomelatine has been proven to be better and well tolerated than SSRIs and SNRIs in
clinical studies. This drug not only mitigates stress, depression, neurochemical
abnormalities, and neuronal atrophy but also normalizes distorted sleep and circa-
dian rhythm. The presence of the MT1 receptor in the dorsal raphe nucleus suggests
their involvement in the pathogenesis of depression (Ng et al. 2017). The knockout
model of MT1 receptor resembles the behavioral changes of depression in experi-
mental animal studies.
The role of melatonin was also evaluated for drug addiction, abuse, and reward. It
is observed via locomotor sensitization, a phenomenon involving repetitive
injections of psychostimulants to induce locomotion in rats during drug free period.
Genetic deletion of MT1 receptor enhances locomotor sensitization in rodents
against methamphetamine (Hutchinson et al. 2012). Hence, the MT1 receptor
could be a novel target to treat the addiction behavior induced by psychostimulants.
Epilepsy
Various studies on the role of epilepsy have revealed that epileptic seizures are
improvised with the help of melatonin but the effect of melatonin as a single
9 Pharmacology of Melatonin and Its Receptors 313
therapeutic moiety is still to be investigated. Due to its ability to cross the blood–
brain barrier it may be used in the treatment of seizures. For example, temporal lobe
epilepsy (TLE) involves progressive development of complex partial seizures that
originate from the temporal lobe (hippocampus) and its various symptoms including
seizures, cognition, and behavioral abnormalities are caused by localized regional
damage in the hippocampus. It includes a gradual decrease of neurogenesis, loss of
GABAergic interneurons, synaptic plasticity, and chronic inflammation which occur
in a bidirectional manner with irregular circadian rhythms. This interaction has been
validated via observing the decreased expression of MT1 receptor in the hippocam-
pus which usually provides an inhibitory effect on the CNS. In this way, agonists of
melatonin receptors could provide an anticonvulsant effect in epilepsy. Both mela-
tonin and agomelatine have been reported to have anticonvulsant action in acute and
chronic preclinical epilepsy. Even the protein and mRNA expression of the binding
site for melatonin named RORα gets decreased in pilocarpine model of epilepsy
(de Alencar Rocha et al. 2017). It has been estimated that RORα may contribute to
temporal epileptic seizures identical to its participation in circadian rhythm, anti-
inflammatory action, and antioxidant properties of melatonin.
Insomnia
High levels of melatonin is observed in adolescence, which declines slowly after the
age of 20. Thus, it affects the age group of 20 more efficiently. Soporific and
hypothermic effects are observed after administration of melatonin during the day
in the young age group. Improvisation in sleep patterns is observed with increased
nocturnal melatonin level via oral administration. Therefore, melatonin therapy is
considered good to treat insomniac conditions.
Depression
It is the property of ideal antidepressant to reduce sleep onset difficulties, without
hindering freshness and daytime alertness. Melatonin does not possess any abuse
potential and adverse effects like “hangover”: it has the ability to improvise patterns
of sleep in patients with insomnia associated with depression.
Alzheimer’s Disease
Accumulation of amyloid β (Aβ) protein and neurofibrillary tangles in the brain is
the main cause of AD. Degeneration takes place as a result of hyper-phosphorylation
of nerve fibers due to age-dependent decline in melatonin. Melatonin plays an
important role in glycogen synthase kinase 3 (GSK-3) modulation; it helps to
prevent neurodegeneration by the influence of AD by the interaction of GSK-3
with presenilin-1, a cofactor for G secretase. It is found that due to its antioxidant
potential, melatonin has direct action in the inhibition of Aβ accumulation and
improving sleep disturbances caused by AD.
Parkinson’s Disease
Oxidative stress is the major factor responsible for the progression of PD; dopamine
metabolism and mitochondrial impairment are the leading causes to generate
314 S. Singh et al.
oxidative stress; these are the major factors responsible for the progression of
PD. Positive therapeutic results are observed when melatonin is used against
rotenone-induced dopamine loss. Due to excessive stimulation of glutamate
receptors, neuronal damage may occur and melatonin has been shown to have a
neuroprotective action against glutamate-induced excitotoxicity. Oxidative stress
and mitochondrial dysfunction in the brains of patients with PD showed elevated
oxidative damage to DNA, decreased levels of glutathione, and increased mono-
amine oxidase activity. Reduced antioxidant defense mechanisms in PD brains are
observed due to a reduction in catalase activity and reduced glutathione (GSH)
(Singh and Jadhav 2014).
Reduced Postganglionic
Melatonin Pineal Sympathetic
Secretion Gland
NE
Dorsal
Cervical
Ganglion
(-)
Retino-
hypothalamic Preganglionic
pathway
SCN
Light Brain
Stem Spinal Cord
Inferior accessory
optic tract
Retina
MT2 receptor. Secondly, the identical receptor has been shown to have analgesic
properties in hot water tail flick test (Yu et al. 2000). The positive result obtained
from the administration of melatonin gets blocked by luzindole and 4P-PDOT which
demonstrate the nociceptive action of MT2 receptor. Besides this, oral administra-
tion of melatonin decreases the flinching behavior associated with tactile allodynia in
diabetic rats (Ambriz-Tututi and Granados-Soto 2007), and this analgesic effect is
reported to get reversed by MT2 receptor antagonist (K-185).
MT3 receptors are abundantly distributed in the peripheral tissue as well as the brain
of hamster. It is believed that phosphoinositide hydrolysis is stimulated by these
receptors. Both melatonin and its precursor (N-acetylserotonin) possess the ability to
activate MT3 receptor, and they also possess pharmacological profile arranged
according to affinities: the sequence includes 2-iodomelatonin > N-acetyl-seroto-
nin > melatonin and the sequence is slightly similar to the human MT1/MT2
receptors (2-iodomelatonin > melatonin > N-acetyl-serotonin). Specific ligands
for the MT3 melatonin receptor are N-acetyltryptamine and prazosin (Oxenkrug
2005). According to suggested hypothesis, mammalian MT3 site radioligand
2-[125I]-MCA-NAT binds to an enzyme quinone reductase 2 in the kidney mem-
brane of hamster. The cloning of this enzyme is done followed by its purification
from hamster kidney membranes. Decreasing intraocular pressure and inhibiting
320 S. Singh et al.
9.6.3.2 Depression
Melatonin and its immediate precursor N-acetylserotonin (NAS) have been reported
to possess antidepressant action in preclinical and clinical studies. Further different
studies have evaluated the role of selective agonist and antagonist of MT3 receptor in
depression studies which include 5-methoxycarbonylamino-N-acetyltryptamine
(5MCA-NAT) and prazosin, respectively (Oxenkrug et al. 2010). The antidepressant
activity of 5-MCA-NAT has been reported to provide significant results in tail
suspension through QR2/MT3 receptor binding site which reverses via the adminis-
tration MT3 receptor antagonist, prazosin. Another drug like resveratrol is also
proven to be potent antagonists of QR2 binding site of MT3 receptor which may
potentiate via the inhibition of indoleamine 2, 3-dioxygenase, a rate limiting step in
tryptophan metabolism and kynurenine pathway. In other compounds like melatonin
and 5-MCA-NAT, this effect may be achieved through competitive inhibition of
tryptophan 2, 3-dioxygenase. In this way, the protective effect of melatonin via MT3
receptor may provide more reliable results in future preclinical and clinical studies.
9 Pharmacology of Melatonin and Its Receptors 321
9.7 Conclusion
Melatonin is a highly significant moiety, which is not only responsible for inducing
sleep but also contributes in a versatile manner to control and coordinate the different
biological processes of the body which includes maintenance of homeostasis,
secretion of essential hormones, bowel movement, biological clock, circadian
rhythm, and many more. MT1 and MT2 are the two important receptor types, and
its subtypes are located at different regions of the body such as skin, parotid gland,
colon, duodenal enterocytes, platelets, white and brown adipocytes, kidney, cells of
the immune system, ovary/granulosa cells, placenta, and myometrium. These
receptors are generally GPCR type, playing a major role at ground level to achieve
various biological goals. Therefore, this luminary molecule is a crucial component
which directs the smooth biological functioning of the human body.
References
Al-Ghoul WM, Herman MD, Dubocovich ML (1998) Melatonin receptor subtype expression in
human cerebellum. Neuroreport 9(18):4063–4068. LWW
Ambriz-Tututi M, Granados-Soto V (2007) Oral and spinal melatonin reduces tactile allodynia in
rats via activation of MT2 and opioid receptors. Pain 132(3):273–280. Elsevier
Arendt J, Deacon S (1997) Treatment of circadian rhythm disorders–melatonin. Chronobiol Int
14(2):185–204. Taylor & Francis
Asghari MH et al (2017) Melatonin as a multifunctional anti-cancer molecule: implications in
gastric cancer. Life 185:38–45. Elsevier
Aust S et al (2004) The melatonin receptor subtype MT1 is expressed in human gallbladder
epithelia. J Pineal Res 36(1):43–48. Wiley Online Library
Ayoub MA et al (2004) Preferential formation of MT1/MT2 melatonin receptor heterodimers with
distinct ligand interaction properties compared with MT2 homodimers. Mol Pharmacol 66
(2):312–321. ASPET
Bahna SG et al (2014) Regional upregulation of hippocampal melatonin MT2 receptors by valproic
acid: therapeutic implications for Alzheimer’s disease. Neurosci Lett 576:84–87. Elsevier
Baltatu OC et al (2017) Melatonin, mitochondria and hypertension. Cell Mol Life Sci
74(21):3955–3964. Springer
Cajochen C et al (2002) Role of melatonin in the regulation of human circadian rhythms and sleep.
9th Triennial Meeting of the European-Pineal-and-Biological-Rhythm-Society, 15, pp 432–437
Cardinali DP et al (2012) Melatonin and its analogs in insomnia and depression. J Pineal Res 52
(4):365–375. Wiley Online Library
Cecon E, Oishi A, Jockers R (2018) Melatonin receptors: molecular pharmacology and signalling in
the context of system bias. Br J Pharmacol 175(16):3263–3280. Wiley Online Library
Comai S, Gobbi G (2014) Unveiling the role of melatonin MT2 receptors in sleep, anxiety and other
neuropsychiatric diseases: a novel target in psychopharmacology. J Psychiatry Neurosci 39
(1):6–21
Das A et al (2013) Overexpression of melatonin membrane receptors increases calcium-binding
proteins and protects VSC4. 1 motoneurons from glutamate toxicity through multiple
mechanisms. J Pineal Res 54(1):58–68. Wiley Online Library
Dawoodi Z, Shah N, De Sousa A (2012) Melatonin agonists: A brief clinical review. Delhi
Psychiatry J 15:268–273. Citeseer
de Alencar Rocha AKA et al (2017) Altered MT1 and MT2 melatonin receptors expression in the
hippocampus of pilocarpine-induced epileptic rats. Epilepsy Behav 71:23–34. Elsevier
322 S. Singh et al.
Abstract
P. K. Deb (*)
Department of Pharmaceutical Sciences, Faculty of Pharmacy, Philadelphia University, Amman,
Jordan
e-mail: [email protected]
S. F. Kokaz · S. N. Abed · B. Chandrasekaran · W. Hourani · A. Y. Jaber
Faculty of Pharmacy, Philadelphia University, Amman, Jordan
R. P. Mailavaram
Department of Pharmaceutical Chemistry, Shri Vishnu College of Pharmacy, Vishnupur (Affiliated
to Andhra University), Bhimavaram, W.G. Dist., AP, India
P. Kumar
Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical
University, Bathinda, Punjab, India
K. N. Venugopala
Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-
Ahsa, Kingdom of Saudi Arabia
Department of Biotechnology and Food Technology, Durban University of Technology, Durban,
South Africa
Keywords
Abbreviations
AC Adenylyl cyclase
ADA Adenosine deaminase
AK Adenosine kinase
AMP Adenosine monophosphate
AR Adenosine receptor
ARNO ADP ribosylation factor nucleotide site opener
ATP Adenosine triphosphate
BBB Blood-brain barrier
CADD Computer-aided drug design
cAMP Cyclic adenosine monophosphate
CNT Concentrative nucleoside transporter
COPD Chronic obstructive pulmonary disease
CREB c-AMP-responsive element binding protein
DAG Diacylglycerol
ENT Equilibrative nucleoside transporter
ERK Extracellular signal-regulated kinase
GPCR G-protein-coupled receptor
GSK-3β Glycogen synthase kinase-3β
HFpEF Heart failure with preserved ejection fraction
iNKT cells Invariant natural killer T cells
iNOS Inducible nitric oxide synthase
IP3 Inositol 1,4,5-triphosphate
IR Ischemia-reperfusion
JNK c-Jun N-terminal kinase
LBDD Ligand-based drug design
MAPK Mitogen-activated protein kinase
MPI Myocardial perfusion imaging
OHT Orthotopic heart transplantation
PAM Positive allosteric modulator
PD Parkinson’s disease
PDEs Phosphodiesterases
PKA Protein kinase A
PKC Protein kinase C
10 Pharmacology of Adenosine Receptors 327
PLC Phospholipase C
PLD Phospholipase D
SAHH S-adenosyl-homocysteine hydrolase
SAMe S-adenosylmethionine
SBDD Structure-based drug design
SPECT Single photon emission computed tomography
TNFα Tumor necrosis factor-alpha
TRAX Translin-associated protein X
US FDA United States Food and Drug Administration
USP4 Ubiquitin-specific protease
10.1 Introduction
O A1 AR Treatment of asthma
H antagonist
H 3C N
N
O N N
O A1 AR Treatment of asthma
O antagonist
H3C O
N N
O N N
Doxofylline (3) CH3
OH A1 AR Treatment of asthma
H3C antagonist
N
O
H 3C N
N
O N N
CH3
Bamifylline (4)
H3C NH N
N N
O N O
HO
Regadenoson (5) HO
HO
O N N O
O
Istradefylline (6)
All the four subtypes of ARs present common molecular structure arrangement,
composed of seven transmembrane helices (TMs 1–7) which are connected to each
other through three intracellular loops (ILs 1–3) and three extracellular loops (ELs
1–3) of varying lengths and functions. These three ELs play important roles in
mediating receptor functions, where cysteine residues connect these ELs by forming
disulfide bonds. The N-terminal containing glycosylation site is present on the
10 Pharmacology of Adenosine Receptors 331
cAMP
ecto-PDE
CD39 CD39 CD73 ADA
ATP ADP 5’-AMP Adenosine Inosine
ENT CNT
Extracellular
Inosine
10.4.1 Distribution of A1 AR
A1AR has shown a high abundance in the brain as well as other organs and tissues.
This receptor subtype has been demonstrated by radioligand-receptor binding stud-
ies and imaging (Elmenhorst et al. 2012; Hayashi et al. 2017), along with RNA
expression, Western blot, as well as functional characterization. Therefore, the wide
distribution of this receptor has suggested its important physiological roles including
spanning neurotransmitter release, neuronal excitability dampening, sleep/wakeful-
ness control, reduction of pain, along with the sedative, anxiolytic, anticonvulsant, as
well as locomotor depressant effects (Gessi et al. 2011; Sawynok 2016). In the
central nervous system (CNS), A1AR is mainly expressed in the brain cortex,
hippocampus, cerebellum, spinal cord, autonomic nerve terminals, and glial cells
(Ballesteros-yáñez et al. 2018; Chen et al. 2013). In the heart, the expression of
A1AR has been shown to be higher in atria and much less in the ventricular
myocardium (Stenberg et al. 2003; Varani et al. 2017). At the vascular level,
A1ARs are found on the coronary smooth muscle arteries as well as endothelial
cells (Headrick et al. 2013). Moreover, A1ARs have been detected in the endothelial
cells of the lung, in the airway’s smooth muscles, in the alveolar epithelial cells, and
in immune cells such as macrophages, neutrophils, eosinophils, and monocytes
(Boros et al. 2016; Sachdeva and Gupta 2013; Sun et al. 2005), where they
essentially promote some proinflammatory effects (Ponnoth et al. 2010). A1AR is
also found in the kidney, adipose tissue, and pancreas, where it causes induction of
negative chronotropic, inotropic, as well as dromotropic effects, reduction in the
renal blood flow and renin release, and inhibition of lipolysis and insulin secretion,
respectively (Dhalla et al. 2009; Prystowsky et al. 2003; Rabadi and Lee 2015; Sun
et al. 2001; Vallon and Mu 2006; Vincenzi et al. 2012). In the kidney, A1ARs mostly
present in the papilla’s collecting ducts, inner medulla, in addition to the cells of the
juxtaglomerular apparatus. A1ARs have been also detected in the retina, skeletal
muscle, intestine, and vascular cells of skeletal muscle (Soni et al. 2017; Varani et al.
2017).
well as in other different cell types such as fibroblasts, smooth muscles, alveolar
epithelial, chromaffin, and taste cells, platelets, myocardial cells, and retinal, intesti-
nal, endothelial and pulmonary epithelial cells (Aherne et al. 2011).
It has been shown in recent development of A2BAR-knockout/lacZ-knocking
mice (Yang et al. 2006) that A2B AR has a wide distribution in numerous tissues
and organs, and this includes the aortic vascular smooth muscle, vasculature, cecum,
brain, large intestine, and urinary bladder (Wang and Huxley 2006; Yaar et al. 2005).
Moreover, A2B AR was found to be highly expressed in various cell types, including
several immune cells such as mast cells (Hua et al. 2007; Yang et al. 2006),
neutrophils (Ryzhov et al. 2008), dendritic cells (Addi et al. 2008), macrophages
(Novitskiy et al. 2008), as well as lymphocytes (Yang et al. 2006), in addition to
other cell types that include the type II alveolar epithelial cells (Eckle et al. 2008),
endothelial cells (Cagnina et al. 2009), chromaffin cells (Yang et al. 2006),
astrocytes (Peakman and Hill 1994), neurons (Christofi et al. 2001), and taste cells
(Stein et al. 2001).
10.4.3 Distribution of A3 AR
The identification of the A3 AR distribution has been made possible after the
generation of cDNA for this receptor (Nishida et al. 2014). The A3 AR subtype
was found to have wide expression in various primary cells, tissues, as well as cell
lines. In the brain, A3AR has been reported in low levels, where it is expressed
particularly in the hypothalamus, thalamus, hippocampus, cortex, as well as retinal
ganglion cells, and motor nerve terminals, in addition to the pial and intercerebral
arteries (Burnett et al. 2010; Janes et al. 2014). Studies have also shown that the
expression of A3 ARs is also reported in microglia and astrocytes; thus inhibiting the
neuro-inflammatory response in these particular cells was shown to be associated
with the analgesic effect they induce (Borea et al. 2016). Despite the cardio-
protective effects that have been related to the A3 AR, as well as the great expression
of this receptor subtype in the coronary and carotid artery, its precise location in the
heart is not yet reported. At the periphery, A3 AR was found to be expressed in
enteric neurons, epithelial cells, lung parenchyma, colonic mucosa, and bronchi.
Moreover, a broad distribution of A3 AR subtype has been reported in inflammatory
cells (Janes et al. 2014) including mast cells, eosinophils, monocytes, neutrophils,
macrophages, dendritic cells, foam cells, lymphocytes, bone marrow cells,
splenocytes, lymph nodes, chondrocytes, synoviocytes, as well as osteoblasts,
where it is responsible for mediating various anti-inflammatory effects (Borea
et al. 2015). It is worth mentioning that A3 AR subtype is overexpressed in some
cancer cells and tissues, which therefore shows the important antitumoral role of this
receptor subtype (Borea et al. 2016). At cellular level, A3 ARs have shown wide
expression in motor nerve terminals, astrocytes, microglia, cortex, as well as retinal
ganglion cells (Borea et al. 2015; Gessi et al. 2013).
334 P. K. Deb et al.
Numerous signal transduction pathways are triggered by all the four G-protein-
coupled ARs based on the activation of a particular type of cell (Fredholm et al.
2001, 2011).
A1 AR
AC
+ Gi -
PIP2 DAG
ATP cAMP
IP3 PKC
-
+ - P P
PKA
P
P38 JNK
ERK1/2
proliferation PLC, PKC-δ, ERK, JNK, and AKT (Gessi et al. 2017). Signal trans-
duction pathway of A2A AR is depicted in Fig. 10.3.
Similar to the A2A AR subtype, the A2B AR is also coupled to Gs protein, triggering
the AC activity and thereby increasing the cAMP levels, PKA phosphorylation, and
cAMP-dependent recruitment of different effectors like exchange proteins (Epac)
(Fredholm et al. 2011). A2B AR-stimulated activation of Epac was also found to
affect the proliferation of umbilical vascular endothelial cells and induce early gene
expression reducing the proliferation of smooth muscle cells of coronary artery in
humans (Fang and Olah 2007; Mayer et al. 2011). Unlike A2A AR, the A2B AR is
also coupled to Gq protein, stimulating PLC leading to Ca2+ mobilization, while
regulating the ion channels through the recruitment of γ subunits. A2B AR can
regulate various pathophysiological functions in the central and peripheral system
through the activation of MAPK and AKT (Sun and Huang 2016). Additionally, A2B
AR responses can be influenced by its various binding partners like netrin-1,
E3KARP-EZRIN-PKA, SNARE, NF-κB1/P105, and α-actinin-1. In particular, the
neuronal guidance protein netrin-1 can bind and activate A2B AR during hypoxia,
reducing the migration of neutrophils and consequent inflammation (Rosenberger
336 P. K. Deb et al.
A2A AR
AC
Gs Gs +
GTP
GDP ATP cAMP
+
P
+ PKA
P
P38
P
JNK
+ +
ERK1/2 P P
AKT CREB
et al. 2009). SNARE protein can bind and translocate the A2B AR from the
cytoplasm to the plasma membrane following agonist binding (Wang et al. 2004)
and consequently, a multiprotein complex with E3KARP (NHERF2) and ezrin
enables the fixation/stabilization of the A2B AR at the cell surface (Sitaraman et al.
2002). Interestingly, α-actinin-1 can promote the dimerization of A2A and A2B ARs,
inducing the cell surface expression of the later (Moriyama and Sitkovsky 2010).
Furthermore, interaction of P105 with A2B AR has shown to reduce the inflamma-
tory effects of NF-κB (Sun et al. 2012). Recently, it has been reported that the
stimulation of A2B AR reduces ERK1/2, p38, and NF-κB induced by RANKL,
thereby reducing osteoclastogenesis in bone (Kim et al. 2017). Several reports also
indicate the role of A2B AR signaling in neuroinflammation (Koscsó et al. 2012;
Merighi et al. 2017), inflammatory bowel disease (Chin et al. 2012; Dammen et al.
2013), cardiac ischemic preconditioning (Yang et al. 2011), atherosclerosis devel-
opment (Gessi et al. 2010a), and reduction of cardiac fibrosis (Phosri et al. 2018,
2017). The signal transduction pathway of A2B AR is depicted in Fig. 10.4.
AAB AR
AC
Gq Gs +
+ Gs
PIP2 DAG GTP GTP
GDP ATP cAMP
IP3 PKC +
P
+ PKA
P
P
P38 JNK
AKT
ERK1/2
DNA binding capability (Fishman et al. 2012, 2004, 2002; Stemmer et al. 2008). A3
AR facilitated neuro- and cardio-protection is regulated via different signaling
pathways including G-protein RhoA and phospholipase D (PLD) (Borea et al.
2018a, b). A3 AR-mediated anti-inflammatory effects are regulated through
MAPK, PI3/Akt, and NF-kB transduction pathways (Ochaion et al. 2008). A3 AR
is also found to induce ERK1/2 and proliferation of cells in human fetal astrocytes,
microglia, glioblastoma, and melanoma among others (Hammarberg et al. 2003;
Merighi et al. 2007; Neary et al. 1998; Soares et al. 2014). Interestingly, reduced
ERK activation was also evident in melanoma, prostate cancer, and glioma cells,
decreasing the proliferation of cells and release of TNF-α (Hyun et al. 2012; Martin
et al. 2006). Activation of A3 AR also modulates p38 and JNK in various cell types
including cancer cells like colon carcinoma (Gessi et al. 2010b). The signal trans-
duction pathway of A3 AR is depicted in Fig. 10.5.
Readers are also encouraged to read the valuable chapter written by Merigi et al.,
highlighting various research findings showcasing the involvement of AR signaling
in diverse pathophysiological conditions (Merighi et al. 2018).
A3 AR
AC
+ Gi -
PIP2 DAG
ATP cAMP
IP3 PKC P
+
P
-
P PKA
P38 JNK
-
-
ERK1/2
NFkb
+ GSK-3b
PKB
+ - -
Cyclin-D1 - b -catenin
C-Myc
NH2 NH2
NH2
N N N NH
N N
N
N HN CH3 N
N N O N N O
O N Br
O N O O
OH HN OH
OH HO OH
OH OH
OH
Regadenoson (5) NECA (7) MRS 3997 (8)
I I
HN HN
N N N N
N N N N Cl
O OH O
H OH
H N
N
OH OH
O O
10.6.1.1 Regadenoson
Regadenoson (5), a selective A2A adenosine receptor agonist, was approved by the
FDA (Food and Drug Administration) in 2008 in the injection form as a pharmaco-
logic stress agent for patients unable to perform adequate exercise in order to
increase blood flow in coronary arteries for myocardial perfusion imaging (MPI)
10 Pharmacology of Adenosine Receptors 339
10.6.1.2 NECA
In recent years, adenosine receptors have shown to be possible pharmacological
targets to alter BBB integrity. A study included intravenous administration of NECA
(50 -N-ethylcarboxamide adenosine) (6), a nonselective ARs agonist, has resulted in
increasing brain concentration of dextrans (both low molecular weight and high
molecular weight). However, NECA pharmacological effect was dose-specific,
producing highest effect at 0.08 mg/kg; lower or higher doses showed less effect.
It was interpreted that doses higher than 0.08 mg/kg of NECA showed less effect due
to adenosine receptors desensitization. The fact that adenosine receptor agonists can
be found in the market and are clinically approved makes these findings even more
valuable presenting a possible less invasive method for BBB disruption (Carman
et al. 2011; Cheng et al. 2016; Malpass 2011).
NECA intraperitoneal administration has shown to increase fasting serum glucose
level. Further investigation showed that NECA administration has elevated glucose
6-phosphatase (G6Pase) enzyme mRNA leading to an increase in the liver G6Pase
enzyme and gluconeogenesis, which is thought to be the cause for serum glucose
elevation (Matsuda et al. 2014). NECA has also been studied for reducing intestinal
IR injury in rats. Results showed that NECA reduced leukocyte activation and
caused a significant improvement in capillary perfusion, thus reducing intestinal
IR injury (Zhou et al. 2015).
O OH
HN
N O
N
N
N
O
OH N N
C C
O O OH
N Cl
H2N N S
NH
S
CVT-2759 (11) Capadenoson (BAY68-4986) (12)
O
O
N
H
OH O NH2
O O
N N N N
C C C C
N N Cl
N N S Cl N N S
S S
Neladenoson (13) Neladenoson bialanate (BAY 1067197) (14)
10.6.2.3 Neladenoson
Neladenoson, an A1 AR partial agonist (12), currently is being tested clinically on
patients with chronic heart failure in the form of dipeptide prodrug. Neladenoson
shows higher selectivity to A1 AR as compared to capadenoson. Many promising
effects caused by Neladenoson have been observed including improvement in
cardiac function without causing undesired effects on blood pressure, atrioventricu-
lar blocks, or bradycardia. The preference of using a partial agonist instead of full
agonist of A1 AR is due to the fact that partial agonist can activate the receptors
without producing severe adverse effects as compared to full agonists. A multiple
dose phase II study (NCT02040233) of Neladenoson has been also conducted to
investigate tolerability, pharmacokinetics, and safety in patients with chronic heart
failure (ParSiFAL study) (Jacobson et al. 2019; Voors et al. 2017).
O R O
NH2
H3C N
N N N
N N N O
O N N N O O
O N N
CH3 N N
O N
O N
Caffeine (15), R=CH3
Theophylline (2), R=H Istradefylline (6) Preladenant (16)
H3C
NH2 O F
O F
Br H
N F
N N N N
N N O
N N N N
N N
H2 N N N O O N
N N
10.6.3.2 PBF-680
The PBF-680 is an A1 AR potent antagonist (structure not disclosed) that is currently
in clinical trials for the treatment of asthma. An ongoing phase II trial
(NCT02635945) aimed to evaluate the efficacy of PBF-680 in patients with mild
to moderate asthma. In this study, 10 mg of PBF-680 was administered orally for
5 days; the efficacy was evaluated by the amount to attenuation of late asthmatic
responses that occurs due to allergen broncho-provocation. Previous studies have
shown that the activation of adenosine A1 receptors has a pro-inflammatory role in
certain immune cells and also broncho-constrictory effect in pulmonary tissue.
Adenosine on the other hand has shown to provoke bronchoconstriction in asthmatic
patients, while an adenosine receptor antagonist such as theophylline is an effective
drug for asthma treatment. Selective A1 receptor antagonists may offer a promising
therapeutic option for asthmatic patients in the future (Gao and Jacobson 2017).
10.6.3.3 Istradefylline
Istradefylline (15) was the first selective A2A AR antagonist; initially it was available
only in Japan for treating the wearing-off phenomenon in Parkinson’s disease
patients receiving levodopa-containing treatment (Saki et al. 2013).
A recent clinical trial of Istradefylline on 31 patients with Parkinson’s disease has
proven its effect in decreasing gait disorders including slow walking speed, short
steps, forward-bent posture, toe dragging, and reduced arm swing which improved
the quality of life of those patients without a serious adverse effect detected (Iijima
et al. 2019). Istradefylline has also been investigated in clinical trials for improving
mood disorders in PD patients. Doses between 20 and 40 mg of Istradefylline were
administered for 12 weeks. Results have shown an improvement in overall mood
disorders. However, further trials are needed to confirm the effectiveness of
istradefylline due to the fact that this trial recruited only 30 patients with dropout
rate of 17% and it was an open-label trial which indicates the possibility of placebo
effect in patients (Nagayama et al. 2019). Recently, it has got the US FDA approval
(2019) and available in the market as an add-on to levodopa/carbidopa for the
treatment of PD (Hoffman 2019; Voelker 2019).
10.6.3.4 Preladenant
Preladenant (16) is an A2A AR antagonist; mainly it was developed to treat patients
with PD. However, clinical trials have not been successful and got discontinued. The
development of preladenant was discontinued in 2013 after two phase III clinical
trials to test its efficacy in treating fluctuating motor disturbances in patients. Results
indicated that preladenant had no significant effect as compared to placebo (Pinna
et al. 2018).
A preladenant phase I study (NCT03099161) in combination with
pembrolizumab was conducted to treat neoplasm. Solid tumors that do not respond
to conventional therapy were targeted in the trial. The study was to assess the
efficacy and safety of preladenant as a treatment and to set the recommended dose
for further clinical trials. However, the study was terminated because the data did not
support the study end point (Congreve et al. 2018).
346 P. K. Deb et al.
10.6.3.5 PBF-509
PBF-509 (17) is a non-xanthine potent A2A AR antagonist that has been tested for
the treatment of PD on rodent models. Studies have shown its efficacy in reducing
pilocarpine-induced tremulous jaw movements, haloperidol-mediated catalepsy, and
L-DOPA-induced dyskinesia, which indicates that PBF-509 is an anti-dyskinetic
agent along with reversing parkinsonian motor impairments making it a potential
treatment option for PD in the future (Núñez et al. 2018).
10.6.3.6 CPI-444
CPI-444 (18) is a selective and highly potent A2A AR antagonist for oral adminis-
tration. The adenosine A2A receptors expressed on immune cells have a suppressive
effect on antitumor activity. Blockage of this receptor with a compound such as
CPI-444 has shown to restore IL2 and IFNγ production and T-cell signaling in
in vitro studies. Preclinical studies of CPI-444 on mice have proven its efficacy in
producing antitumor response when anti–PD-L1 immunotherapy failed to produce
the required therapeutic response. The mechanism that explains how blocking of
A2A receptors can overcome the resistance of anti–PD-L1 treatment is still under
investigation (Willingham et al. 2018).
A clinical phase I trial (NCT02655822) is currently ongoing (by Corvus
Pharmaceuticals, Inc.) for dose selection, tolerability, and safety of CPI-444 as a
single antitumor agent or in combination with atezolizumab. Adenosine has shown
to suppress antitumor activity in immune cells (T-cells) (Mobasher et al. 2019).
O
R O
N HN
HN HN
NH
H3 C
N OCH3 N N
O N NH N NH
N
S NH2 H
N
neuropathic pain and inflammatory models. It was also noticed to induce sedation
after the initial dosing; 5 days after daily administration tolerance has occurred due to
downregulation of the A1 AR. T62 has progressed into clinical trials, a phase II trial
(NCT00809679) to evaluate the safety and efficacy of this compound as an analgesic
for patients with postherpetic neuralgia. However, some patients experienced tran-
sient elevations in liver enzymes (transaminases) which terminated the study
(Romagnoli et al. 2015; Sawynok 2016). LUF 5484 (2-amino-4,5,6,7-
tetrahydrobenzo[b]thiophen-3-yl)(3,4-dichlorophenyl)methanone (21) is an A1
adenosine receptor allosteric modulator (Bueters et al. 2002).
10.6.4.2 VUF5455
VUF5455 (22) is a 3-(2-pyridinyl) isoquinoline derivative, the first selective PAM of
A3 AR. VUF5455 enhances the binding of A3 receptor agonists and increases the
dissociation rate of antagonist (Briddon et al. 2018; Soudijn et al. 2006).
10.6.4.3 LUF6000
LUF6000 (2-Cyclohexyl-N-(3,4-dichlorophenyl)-1H-imidazo[4,5-c]quinolin-4-
amine) (23), an A3 AR PAM, increases the activity of orthosteric agonists. The
maximal effect of the native ligand increases by 45% when an allosteric enhancer
binds to the receptor. LUF6000 has been studied on animal models including mice
and rats, and results have shown that LUF6000 induces anti-inflammatory effect by
slightly stimulating neutrophils and normal white blood cells (Cohen et al. 2014).
10.6.4.4 LUF6096
LUF6096 (N-{2-[(3,4-dichlorophenyl)amino]quinolin-4-yl}cyclohexanecarbox-
amide) (24) is a positive A3 AR allosteric modulator; it was developed by the
scission of the imidazole ring of LUF6000. LUF6096 has been through preclinical
studies on animal models and human cell membranes to evaluate its efficacy in
reducing myocardial ischemia/reperfusion injury. Results have shown that LUF6096
is well tolerated and effective in deceasing the myocardial ischemia/reperfusion
injury on dog models (Du et al. 2018, 2012).
348 P. K. Deb et al.
10.6.4.5 DU124183
DU124183 (2-cyclopentyl-4-phenylamino-1H-imidazo[4,5-c]quinoline) (25) is a
selective allosteric modulator that enhances agonist binding and function of A3
AR (Göblyös and Ijzerman 2009). DU124183 causes a decrease in agonist potency
meanwhile enhancing its maximum effect (Emax) (Gao et al. 2008).
10.7 Conclusions
Adenosine and its four receptor subtypes (A1, A2A, A2B, and A3 ARs) are widely
distributed throughout the body, modulating the physiological and pathological
conditions of almost every organs and tissues. The ubiquitous distribution of ARs
not only signifies their potential drug targets but also imposed a great challenge in
the process of discovery and development of drugs selectively targeting a particular
subtype of AR in disease-specific tissues, while culminating in undesirable side
effects. In the last three decades, extensive research efforts from academia and
pharmaceutical industries resulted in the discovery of various potential ligands
targeting ARs, but only few of them could sustain the clinical trials to successfully
reach the market. Istradefylline, an A2A selective antagonist, is the most recently US
FDA approved (2019) drug available in the market as an add-on to levodopa/
carbidopa for the treatment of PD. Moreover, the recent discovery of the 3D crystal
structure of A1 AR and the previously identified 3D structure of A2A AR have not
only enhanced the understanding of the binding site topology of these receptors but
also facilitated the development of improved homology models of other two AR
subtypes as well as computer-aided structure-based strategies to design and discover
novel AR-specific ligands. In this regard, the future discovery of the 3D crystal
structures of remaining A2B and A3 ARs would further provide a clear insight into all
the four subtypes of ARs, thus boost up the rational drug discovery process and
development of novel clinical candidates, selectively targeting a particular AR
subtype relevant to the therapeutic intervention of specific pathological disorders.
References
Adachi H, Palaniappan KK, Ivanov AA, Bergman N, Gao Z, Jacobson KA (2007) Structure-activity
relationships of 2,N6,50 -substituted adenosine derivatives with potent activity at the A2B
adenosine receptor. J Med Chem 50:1810–1827
Addi AB, Lefort A, Hua X, Libert F, Communi D, Ledent C, Macours P, Tilley SL, Boeynaems J,
Robaye B (2008) Modulation of murine dendritic cell function by adenine nucleotides and
adenosine: involvement of the A 2B receptor. Eur J Immunol 38:1610–1620
Agrawal N, Chandrasekaran B, Al-Aboudi A (2019) Recent advances in the in-silico structure-
based and ligand-based approaches for the design and discovery of agonists and antagonists of
A2A adenosine receptor. Curr Pharm Des 25:774–782
Aherne CM, Kewley EM, Eltzschig HK (2011) The resurgence of A2B adenosine receptor
signaling. Biochim Biophys Acta 1808:1329–1339. https://doi.org/10.1016/j.bbamem.2010.
05.016
10 Pharmacology of Adenosine Receptors 349
Al-attraqchi OHA, Attimarad M, Venugopala KN, Nair A, Noor HA (2019) Adenosine A2A
receptor as a potential drug target - current status and future perspectives. Curr Pharm Des
25:2716–2740
Albrecht-küpper BE, Leineweber K, Nell PG (2012) Partial adenosine A1 receptor agonists for
cardiovascular therapies. Purinergic Signal 8:91–99
Al-Shar’i Nizar A, Al-Balas QA (2019) Molecular dynamics simulations of adenosine receptors:
advances, applications and trends. Curr Pharm Des 25:783–816
Andrikopoulou E, Hage FG (2018) Adverse effects associated with regadenoson myocardial
perfusion imaging. J Nucl Cardiol 25:1724–1731. https://doi.org/10.1007/s12350-018-1218-7
Bading B, Kelm M, Schrader J (1993) Formation and salvage of adenosine by macrovascular
endothelial cells. Am J Physiol Circ Physiol 264:H692–H700
Ballesteros-yáñez I, Castillo CA, Merighi S (2018) The role of adenosine receptors in
psychostimulant addiction. Front Pharmacol 8:1–18
Baltos J, Vecchio EA, Harris MA, Xue C, Ritchie RH, Christopoulos A, White PJ, May LT (2017)
Capadenoson, a clinically trialed partial adenosine A1 receptor agonist, can stimulate adenosine
A2B receptor biased agonism. Biochem Pharmacol 135:79–89. https://doi.org/10.1016/j.bcp.
2017.03.014
Baraldi PG, Tabrizi MA, Gessi S, Borea PA (2008) Adenosine receptor antagonists: translating
medicinal chemistry and pharmacology into clinical utility. Chem Rev 108:238–263
Baratloo A, Negida A, El Ashal G, Behnaz N (2015) Intravenous caffeine for the treatment of acute
migraine: a pilot study. J Caffeine Res 5:1–5
Baratloo A, Rouhipour A, Forouzanfar MM, Safari S, Amiri M, Negida A (2016) The role of
caffeine in pain management: a brief literature review. Anesth Pain Med 6:e33193
Barnes PJ (2013) Theophylline. Am J Respir Crit Care Med 188:901–906
Basheer R, Arrigoni E, Thatte HS, Greene RW, Ambudkar IS, McCarley RW (2002) Adenosine
induces inositol 1,4,5-trisphosphate receptor-mediated mobilization of intracellular calcium
stores in basal forebrain cholinergic neurons. J Neurosci 22:7680–7686
Basu S, Barawkar DA, Ramdas V, Waman Y, Patel M, Panmand A, Kumar S, Thorat S, Bonagiri R,
Jadhav D, Mukhopadhyay P, Prasad V, Reddy BS, Goswami A, Chaturvedi S, Menon S,
Quraishi A, Ghosh I, Dusange S, Paliwal S, Kulkarni A, Karande V, Thakre R, Bedse G,
Rouduri S, Gundu J, Palle VP, Chugh A, Mookhtiar KA (2016) A2B adenosine receptor
antagonists: design, synthesis and biological evaluation of novel xanthine derivatives. Eur J
Med Chem 127:986–996. https://doi.org/10.1016/j.ejmech.2016.11.007
Biber K, Klotz K, Berger M, Gebicke-ha PJ, Van Calker D (1997) Adenosine A1 receptor-mediated
activation of phospholipase C in cultured astrocytes depends on the level of receptor expression.
J Neurosci 17:4956–4964
Bjursell MK, Blom HJ, Cayuela JA, Engvall ML, Lesko N, Halldin M, Falkenberg M, Jakobs C,
Balasubramaniam S, Gustafsson CM, Lundeberg J, Smith D, Struys E, Von Do U, Wedell A
(2011) Adenosine kinase deficiency disrupts the methionine cycle and causes
hypermethioninemia, encephalopathy, and abnormal liver function. Am J Hum Genet
89:507–515
Boison D (2018) Regulation of extracellular adenosine. In: Borea P., Varani, K., Gessi, S., Merighi,
S., Vincenzi, F., (Eds.), The adenosine receptors. pp. 13–32
Boison D, Stone R, Neurobiology D (2013) Adenosine kinase: exploitation for therapeutic gain.
Pharmacol Rev 65:906–943
Borah P, Deka S, Mailavaram RP, Deb PK (2019) P1 receptor agonists/antagonists in clinical trials -
potential drug candidates of the future. Curr Pharm Des 25:2792–2807
Borea PA, Varani K, Vincenzi F, Baraldi PG, Tabrizi MA, Merighi S, Gessi S (2015) The A3
adenosine receptor: history and perspectives. Pharmacol Rev 67(1:74–102.
Borea PA, Gessi S, Merighi S, Varani K (2016) Adenosine as a multi-signalling guardian angel in
human diseases: when, where and how does it exert its protective effects? Trends Pharmacol Sci
37:419–434. https://doi.org/10.1016/j.tips.2016.02.006
350 P. K. Deb et al.
Borea P, Varani K, Gessi S, Merighi S, Vincenzi F (2018a) The adenosine receptors, vol 2018.
Springer, New York, NY
Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K (2018b) Pharmacology of adenosine receptors:
the state of the art. Am Physiol Soc 98:1591–1625
Boros D, Thompson J, Larson DF (2016) Adenosine regulation of the immune response initiated by
ischemia reperfusion injury. Perfusion 31:103–110
Briddon SJ, Kilpatrick LE, Hill SJ (2018) Studying GPCR pharmacology in membrane
microdomains: fluorescence correlation spectroscopy comes of Age. Trends Pharmacol Sci
39:158–174. https://doi.org/10.1016/j.tips.2017.11.004
Brugarolas M, Navarro G, Mart E, Angelats E, Casad V, Lanciego L, Franco R (2014) G-protein-
coupled receptor heteromers as key players in the molecular architecture of the central nervous
system. CNS Neurosci Ther 20:703–709
Bueters TJH, Van Helden HPM, Danhof M, Ijzerman AP (2002) Effects of the adenosine A1
receptor allosteric modulators PD 81,723 and LUF 5484 on the striatal acetylcholine release.
Eur J Pharmacol. 454:177–182
Burnett LA, Blais EM, Unadkat JD, Hille B, Tilley SL, Babcock DF (2010) Testicular expression of
Adora3i2 in Adora3 knockout mice reveals a role of mouse A3Ri2 and human A3Ri3 adenosine
receptors in sperm . J Biol Chem 285:33662–33670
Cagnina RE, Ramos SI, Marshall MA, Wang G, Frazier CR, Linden J, Re C, Si R, Ma M, Wang G,
Cr F (2009) Adenosine A2B receptors are highly expressed on murine type II alveolar epithelial
cells. Am J Physiol Lung Cell Mol Physiol 297:467–475
Carman AJ, Mills JH, Krenz A, Kim D, Bynoe MS (2011) Adenosine receptor signaling modulates
permeability of the blood – brain barrier. J Neurosci 31:13272–13280
Chandrasekaran B, Samarneh S, Jaber A, Kassab G, Agrawal N (2019) Therapeutic potentials of
A2B adenosine receptor ligands: current status and perspectives. Curr Pharm Des 25:2741–2771
Chen X, Ghribi O, Geiger JD (2010) Caffeine protects against disruptions of the blood-brain barrier
in animal models of Alzheimer’s and Parkinson’s diseases. J Alzheimers Dis 20:S127–S141
Chen J, Eltzschig HK, Fredholm BB (2013) Adenosine receptors as drug targets — what are the
challenges? Nat Rev Drug Discov 12:265
Cheng C, Yang YL, Liao KH, Lai TW (2016) Adenosine receptor agonist NECA increases cerebral
extravasation of fluorescein and low molecular weight dextran independent of blood-brain
barrier modulation. Sci Rep 6:1–9. https://doi.org/10.1038/srep23882
Cheng RKY, Segala E, Robertson N, Marshall FH, Cooke RM, Errey JC, Marshall FH, Cooke RM
(2017) Structures of human A1 and A2A adenosine receptors with xanthines reveal
determinants of selectivity. Structure 25:1275–1285
Chin A, Svejda B, Gustafsson BI, Granlund AB, Sandvik AK, Timberlake A, Sumpio B,
Pfragner R, Modlin IM, Kidd M, Chin A, Svejda B, Bi G, Ab G, Ak S, Timberlake A,
Sumpio B, Pfragner R, Im M, The KM (2012) The role of mechanical forces and adenosine
in the regulation of intestinal enterochromaffin cell serotonin secretion. Am J Physiol
Gastrointest Liver Physiol 4:397–405
Christofi FL, Zhang H, Yu J, Guzman J (2001) Differential gene expression of adenosine A1, A2a,
A2b, and A3 receptors in the human enteric nervous system. J Comp Neurol 439:46–64
Cohen S, Barer F, Bar-yehuda S, Ijzerman AP, Jacobson KA, Fishman P (2014) A3 adenosine
receptor allosteric modulator induces an anti-inflammatory effect: in vivo studies and molecular
mechanism of action. Mediators Inflamm 2014:1–8
Cohen S, Barer F, Itzhak I, Silverman MH, Fishman P (2018) Inhibition of IL-17 and IL-23 in
human keratinocytes by the A3 adenosine receptor agonist piclidenoson. J Immunol Res
2018:1–9
Congreve M, Brown GA, Borodovsky A, Lamb ML, Congreve M, Brown GA, Borodovsky A,
Lamb ML, Congreve M, Brown GA, Borodovsky A, Lamb ML (2018) Expert opinion on drug
discovery targeting adenosine A2A receptor antagonism for treatment of cancer. Expert Opin
Drug Discov 13:1–7. https://doi.org/10.1080/17460441.2018.1534825
10 Pharmacology of Adenosine Receptors 351
Dammen R, Haugen M, Svejda B, Alaimo D, Brenna O, Pfragner R, Gustafsson BI, Kidd M (2013)
Correction: the stimulatory adenosine receptor ADORA2B regulates serotonin (5-HT) synthesis
and release in oxygen-depleted EC cells in inflammatory bowel disease. PLoS One 8:e62607
Deb PK (2019a) Therapeutic potentials of adenosine receptors: the state of the art. Curr Pharm Des
25:2789–2791
Deb PK (2019b) Progress in the development of agonists, antagonists and allosteric modulators of
adenosine receptors. Curr Pharm Des 25:2695–2696
Deb PK (2019c) Recent updates in the computer aided drug design strategies for the discovery of
agonists and antagonists of adenosine receptors. Curr Pharm Des 25:747–749
Deb PK, Al-attraqchi O, Al-qattan MN, Prasad MR, Tekade RK (2018a) Applications of computers
in pharmaceutical product formulation, dosage form design parameters. Elsevier, Amsterdam.
https://doi.org/10.1016/B978-0-12-814421-3.00019-1
Deb PK, Mailavaram R, Chandrasekaran B, Kaki VR, Kaur R, Kachler S, Klotz K-N, Akkinepally
RR (2018b) Synthesis, adenosine receptor binding and molecular modeling studies of novel
Thieno[2,3-d]pyrimidine derivatives. Chem Biol Drug Des 91:962–969
Deb PK, Chandrasekaran B, Mailavaram R, Tekade RK, Muttaleb A, Jaber Y (2019a) Molecular
modeling approaches for the discovery of adenosine A2B receptor antagonists: current status
and future perspectives. Drug Discov Today 24:1854–1864. https://doi.org/10.1016/j.drudis.
2019.05.011
Deb PK, Deka S, Borah P, Abed SN, Klotz K (2019b) Medicinal chemistry and therapeutic
potential of agonists, antagonists and allosteric modulators of A1 adenosine receptor: current
status and perspectives. Curr Pharm Des 25:2697–2715
Deussen A (2000) Metabolic flux rates of adenosine in the heart. Naunyn Schmiedebergs Arch
Pharmacol 362:351–352
Deussen A, Stappert M, Schafer S, Kelm M (1999) Quantification of extracellular and intracellular
adenosine production understanding the transmembranous concentration gradient. Circulation
99:2041–2047
Dhalla AK, Chisholm JW, Reaven GM (2009) A 1 adenosine receptor: role in diabetes and obesity.
Handb Exp Pharmacol 193:271–295
Dinh W, Albrecht-k B, Gheorghiade M, Voors AA, Van Der Laan M, Sabbah HN (2017) Partial
adenosine A1 agonist in heart failure. In: Bauersachs J, Butler J, Sandner P (eds) Handbook of
experimental pharmacology, vol 243. Springer, New York, NY, pp 177–203
Du L, Gao Z, Nithipatikom K, Ijzerman AP, Van Veldhoven JPD, Jacobson KA, Gross GJ,
Auchampach JA (2012) Protection from myocardial ischemia/reperfusion injury by a positive
allosteric modulator of the A3 adenosine receptor. JPET 340:210–217
Du L, Gao Z, Paoletta S, Wan TC, Gizewski ET, Barbour S, Van Veldhoven JPD, Ijzerman AP
(2018) Species differences and mechanism of action of A3 adenosine receptor allosteric
modulators. Purinergic Signal 14:59–71
Eckle T, Faigle M, Grenz A, Laucher S, Thompson LF, Eltzschig HK (2008) A2B adenosine
receptor dampens hypoxia-induced vascular leak. Blood 111:2024–2036
Ellis JC, Catherine A, Jones C, Flint J (1995) Activating transcription factor-1 is a specific
antagonist of the cyclic adenosine 3’.5’-monophosphate (cAMP) response element-binding
protein-1-mediated response to cAMP. Mol Endocrinol 9:255–265
Elmenhorst D, Meyer PT, Matusch A, Winz OH, Bauer A (2012) Caffeine occupancy of human
cerebral A1 adenosine receptors: in vivo quantification with 18F-CPFPX and PET. J Nucl Med
53:1723–1730
Fang Y, Olah ME (2007) Cyclic AMP-dependent, protein kinase A-independent activation of
extracellular signal-regulated kinase 1/2 following adenosine receptor stimulation in human
umbilical vein endothelial cells: role of exchange protein activated by cAMP 1 (Epac1). J
Pharmacol Exp Ther 322:1189–1200
Fellner C (2016) More biologic therapies expected to treat advanced plaque psoriasis. Pharm Ther
41:388–390
352 P. K. Deb et al.
Ferre S, Navarro G, Casado V, Cortes A, Mallol J, Canela EI, Lluıs C, Franco R (2010) G protein-
coupled receptor heteromers as new targets for drug development I. Prog Mol Biol Transl Sci
91:41–52
Field JJ, Majerus E, Gordeuk VR, Gowhari M, Hoppe C, Heeney MM, Achebe M, George A,
Chu H, Sheehan B, Puligandla M, Neuberg D, Lin G, Linden J, Nathan DG (2019) Randomized
phase 2 trial of regadenoson for treatment of acute vaso-occlusive crises in sickle cell disease.
Blood Adv 1:1645–1649
Finkelstein JD (1998) The metabolism of homocysteine: pathways and regulation. Eur J Pediatr
157 Suppl:40–44
Fishman P, Cohen S (2016) The A3 adenosine receptor (A3AR): therapeutic target and predictive
biological marker in rheumatoid arthritis. Clin Rheumatol 35:2359–2362
Fishman P, Bar-yehuda S, Madi L, Cohn I, Tikva P, Biopharma IC, Tikva P (2002) A3 adenosine
receptor as a target for cancer therapy. Anticancer Drugs 13:437–443
Fishman P, Bar-yehuda S, Ohana G, Barer F, Ochaion A, Erlanger A, Madi L (2004) An agonist to
the A3 adenosine receptor inhibits colon carcinoma growth in mice via modulation of GSK-3b
and NF-jB. Oncogen 23:2465–2471
Fishman P, Bar-yehuda S, Liang BT, Jacobson KA (2012) Pharmacological and therapeutic effects
of A 3 adenosine receptor agonists. Drug Discov Today 17:359–366. https://doi.org/10.1016/j.
drudis.2011.10.007
Fredholm BB (2014) Adenosine—a physiological or pathophysiological agent? J Mol Med
92:201–206
Fredholm BB, Arslan G, Halldner L, Schulte G, Wasserman W (2000) Structure and function of
adenosine receptors and their genes. Naunyn Schmiedebergs Arch Pharmacol 362:364–365
Fredholm B, IJzerman A, Jacobson K, Klotz K-N, Linden J (2001) International Union of
Pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacol Rev
53:527–552
Fredholm BB, Ijzerman AP, Jacobson KA, Linden J, Mu CE (2011) International Union of Basic
and Clinical Pharmacology. LXXXI. Nomenclature and classification of adenosine receptors —
an update. Pharmacol Rev 63:1–34
Fresco P, Diniz C, Gonc J (2004) Facilitation of noradrenaline release by activation of adenosine
A2A receptors triggers both phospholipase C and adenylate cyclase pathways in rat tail artery.
Cardiovasc. Res. 63:739–746
Gao Z, Jacobson KA (2017) Purinergic signaling in mast cell degranulation and asthma. Front
Pharmacol 8:947
Gao Z, Ye K, Göblyös A, Ijzerman AP, Jacobson KA (2008) Flexible modulation of agonist
efficacy at the human A3 adenosine receptor by the imidazoquinoline allosteric enhancer
LUF6000. BMC Pharmacol 8:20
Gao Z, Balasubramanian R, Kiselev E, Wei Q (2014) Probing biased/partial agonism at the G
protein-coupled A2B adenosine receptor. Biochem Pharmacol 90:297–306
Gessi S, Fogli E, Sacchetto V, Merighi S, Varani K, Preti D, Leung E, MacLennan S, Borea PA
(2010a) Adenosine Modulates HIF-1α, VEGF, IL-8, and Foam Cell Formation in a Human
Model of Hypoxic Foam Cells. Arteriosclerosis, Thrombosis, and Vascular Biology 30(1):90-
97
Gessi S, Sacchetto V, Fogli E, Merighi S, Varani K, Giovanni P, Aghazadeh M, Leung E,
Maclennan S, Andrea P (2010b) Modulation of metalloproteinase-9 in U87MG glioblastoma
cells by A 3 adenosine receptors. Biochem Pharmacol 79:1483–1495. https://doi.org/10.1016/j.
bcp.2010.01.009
Gessi S, Merighi S, Fazzi D, Stefanelli A, Varani K, Borea PA (2011) Adenosine receptor targeting
in health and disease. Expert Opin Investig Drugs 20:1591–1609
Gessi S, Merighi S, Stefanelli A, Fazzi D, Varani K, Borea PA (2013) A1 and A3 adenosine
receptors inhibit LPS-induced hypoxia-inducible factor-1 accumulation in murine astrocytes.
Pharmacol Res 76:157–170. https://doi.org/10.1016/j.phrs.2013.08.002
10 Pharmacology of Adenosine Receptors 353
Jacobson KA, Tosh DK, Jain S, Gao Z (2019) Historical and current adenosine receptor agonists in
preclinical and clinical development. Front Cell Neurosci 13:1–17
Janes K, Esposito E, Doyle T, Cuzzocrea S, Tosh DK, Jacobson KA, Salvemini D (2014) A3
adenosine receptor agonist prevents the development of paclitaxel-induced neuropathic pain by
modulating spinal glial-restricted redox-dependent signaling pathways. Pain 155:2560–2567.
https://doi.org/10.1016/j.pain.2014.09.016
Kim BH, Oh JH, Lee NK (2017) Molecules and cells the inactivation of ERK1/2, p38 and NF-kB is
involved in the down-regulation of osteoclastogenesis and function by A2B adenosine receptor
stimulation. Mol Cells 40:752–760
Kirsch GE, Codina J, Birnbaumer L, Brown AM (1990) Coupling of ATP-sensitive K+ channels to
A1 receptors by G proteins in rat ventricular myocytes. Am J Physiol 259:H820–H826
Kishore DP, Balakumar C, Rao AR, Pratim P, Roy K (2011) QSAR of adenosine receptor
antagonists: exploring physicochemical derivatives with human adenosine A3 receptor subtype.
Bioorg Med Chem Lett 21:818–823. https://doi.org/10.1016/j.bmcl.2010.11.094
Koscsó B, Csóka B, Selmeczy Z, Himer L, Pacher P, Virág L, Haskó G (2012) Adenosine augments
IL-10 production by microglial cells through an A2B adenosine receptor-mediated process. J
Immunol 188:445–453
Koupenova M, Johnston-cox H, Vezeridis A, Gavras H, Yang D, Zannis V, Ravid K (2012) A2b
adenosine receptor regulates hyperlipidemia and atherosclerosis. Circulation 125:354–363
Kull B, Svenningsson P, Fredholm BB (2000) Adenosine A2A receptors are colocalized with and
activate G olf in rat STRIATUM. Mol Pharmacol 58:771–777
Kunduri SS, Dick GM, Nayeem M, Mustafa SJ (2013) Adenosine A1 receptor signaling inhibits
BK channels through a PKCα-dependent mechanism in mouse aortic smooth muscle. Physiol
Rep 1:1–11. https://doi.org/10.1002/phy2.37
Lazarus JJ, Saleh A, Ghannam M, Aaronson K, Colvin M, Pagani F, Murthy VL, Konerman MC
(2018) Safety of regadenoson positron emission tomography stress testing in orthotopic heart
transplant patients. J Nucl Cardiol:1–6. https://doi.org/10.1007/s12350-018-01466-1
Linden J (2005) Adenosine in tissue protection and tissue regeneration. Mol Pharmacol
67:1385–1387
Liu B, Bing Q, Li S, Han B, Lu J, Baiyun R, Zhang X, Lv Y, Wu H, Zhang Z (2019) Role of - A2B
adenosine receptor-dependent adenosine signaling in multi-walled carbon nanotube-triggered
lung fibrosis in mice. J Nanobiotechnol 17:1–11. https://doi.org/10.1186/s12951-019-0478-y
López-cruz L, Salamone JD, Correa M (2018) Caffeine and selective adenosine receptor
antagonists as new therapeutic tools for the motivational symptoms of depression. Front
Pharmacol 9:1–14
Mahmod Al-Qattan MN, Mordi MN (2019) Molecular basis of modulating adenosine receptors
activities. Curr Pharm Des 25:817–831
Mailavaram R, Al-Attraqchi O, Kar S, Ghosh S (2019) Current status in the design and develop-
ment of agonists and antagonists of adenosine A3 receptor as potential therapeutic agents. Curr
Pharm Des 25:2772–2787
Malpass K (2011) Pharmacology: new methods to permeabilize the blood–brain barrier. Nat Rev
Neurol 7:597. https://doi.org/10.1038/nrneurol.2011.161
Martin L, Pingle SC, Hallam DM, Rybak LP (2006) Activation of the adenosine A3 Receptor in
RAW 264.7 cells inhibits lipopolysaccharide-stimulated tumor necrosis factor-a release by
reducing calcium-dependent activation of nuclear factor-kB and extracellular signal-regulated
kinase 1/2. J Pharmacol Exp Ther 316:71–78
Matsuda K, Horikawa Y, Sasaki Y, Sakata SF (2014) The adenosine receptor agonist 5’-N-
ethylcarboxamide-adenosine increases glucose 6-phosphatase expression and gluconeogenesis.
Pharmacol Pharm 5:19–23
Mayer P, Hinze AV, Harst A, Von Ku I (2011) A2B receptors mediate the induction of early genes
and inhibition of arterial smooth muscle cell proliferation via Epac. Cardiovasc Res 90:148–156
10 Pharmacology of Adenosine Receptors 355
Merighi S, Benini A, Mirandola P, Gessi S, Varani K, Leung E, Maclennan S, Baraldi PG, Borea
PA (2007) Hypoxia inhibits paclitaxel-induced apoptosis through adenosine-mediated phos-
phorylation of bad in glioblastoma cells. Mol Pharmacol 72:162–172
Merighi S, Borea PA, Stefanelli A, Bencivenni S, Castillo CA, Varani K, Gessi S (2015) A 2A and
A 2B adenosine receptors Affect HIF-1 a signaling in activated primary microglial cells. Glia
63:1933–1952
Merighi S, Bencivenni S, Vincenzi F, Varani K, Borea PA, Gessi S (2017) Original article A2B
adenosine receptors stimulate IL-6 production in primary murine microglia through p38 MAPK
kinase pathway. Pharmacol Res 117:9–19. https://doi.org/10.1016/j.phrs.2016.11.024
Merighi S, Gessi S, Borea PA (2018) Adenosine receptors: structure, distribution, and signal
transduction. In: Borea P, Varani K, Gessi S, Merighi S, Vincenzi F (eds) The adenosine
receptors, The receptors, vol 34, pp 33–57
Mobasher M, Miller RA, Kwei L, Strahs D, Das V, Luciano G, Powderly JD, Merchan JR, Barve
MA, LoRusso P, Tripathi A (2019) A phase I/Ib multicenter study to evaluate the humanized
anti-CD73 antibody, CPI-006, as a single agent, in combination with CPI-444, and in combina-
tion with pembrolizumab in adult patients with advanced cancers. J Clin Oncol 37:TPS2646–
TPS2646
Moffatt BA, Stevens YY, Allen MS, Snider JD, Pereira LA, Todorova MI, Summers PS,
Weretilnyk EA, Martin-mccaffrey L, Wagner C (2002) Adenosine kinase deficiency is
associated with developmental abnormalities and reduced transmethylation. Plant Physiol
128:812–821
Mohamed RA, Agha AM, Nassar NN (2016) Role of adenosine A 2A receptor in cerebral ischemia
reperfusion injury: signaling to phosphorylated extracellular signal-regulated protein kinase
(pERK1/2). Neuroscience 314:145–159. https://doi.org/10.1016/j.neuroscience.2015.11.059
Moriyama K, Sitkovsky MV (2010) Adenosine A2A receptor is involved in cell surface expression
of A2B receptor . J Biol Chem 285:39271–39288
Nagayama H, Kano O, Murakami H, Ono K, Hamada M, Toda T, Sengoku R, Shimo Y, Hattori N
(2019) Effect of istradefylline on mood disorders in Parkinson’s disease. J Neurol Sci
396:78–83. https://doi.org/10.1016/j.jns.2018.11.005
Nalli AD, Kumar DP, Al-Shboul O, Mahavadi S, Kuemmerle JF, Grider JR, Murthy KS (2014)
Regulation of Gbci-dependent PLC-b 3 activity in smooth muscle: inhibitory phosphorylation
of PLC- b3 by PKA and PKG and stimulatory phosphorylation of Gai -GTPase-activating
protein RGS2 by PKG. Cell Biochem Biophys 70:867–880
Navarro G, Cordomí A, Zelman-femiak M, Brugarolas M, Moreno E, Aguinaga D, Perez-benito L,
Cortés A, Casadó V, Mallol J, Canela EI, Lluís C, Pardo L, García-sáez AJ, Mccormick PJ,
Franco R (2016) Quaternary structure of a G-protein- coupled receptor heterotetramer in
complex with G i and G s. BMC Biol 14:1–12. https://doi.org/10.1186/s12915-016-0247-4
Neary JT, Mccarthy M, Kang Y, Zuniga S (1998) Mitogenic signaling from P1 and P2 purinergic
receptors to mitogen-activated protein kinase in human fetal astrocyte cultures. Neurosci Lett
242:159–162
Newby AC (1984) Adenosine and the concept of ’ retaliatory metabolites ’. Trends Biochem Sci
9:42–44
Nishida K, Dohi Y, Yamanaka Y (2014) Expression of adenosine A2b receptor in rat type II and III
taste cells. Histochem Cell Biol 141:499–506
Novitskiy SV, Ryzhov S, Zaynagetdinov R, Goldstein AE, Huang Y, Oleg Y, Blackburn MR,
Biaggioni I, Carbone DP, Feoktistov I, Mikhail M, Dc W, Novitskiy SV, Ryzhov S,
Zaynagetdinov R, Goldstein AE, Huang Y, Tikhomirov OY (2008) Adenosine receptors in
regulation of dendritic cell differentiation and function. Blood 112:1822–1831
Núñez F, Taura J, Camacho J, López-cano M, Fernández-dueñas V, Castro N, Castro J, Ciruela F,
Aubert J, Castro J (2018) PBF509, an adenosine A 2A receptor antagonist with efficacy in
rodent models of movement disorders. Front Pharmacol 9:1200
Ochaion A, Bar-yehuda S, Cohen S, Amital H, Jacobson KA, Joshi BV (2008) The A 3 adenosine
receptor agonist CF502 inhibits the PI3K, PKB/Akt and NF-k B signaling pathway in
synoviocytes from rheumatoid arthritis patients and in adjuvant-induced arthritis rats. Biochem
Pharmacol 76:482–494
356 P. K. Deb et al.
Pacheco R, Lejeune M, Climent N, Oliva H, Gatell JM, Gallart T, Mallol J (2005) CD26, adenosine
deaminase, and adenosine receptors mediate costimulatory signals in the immunological syn-
apse. PNAS 102:9583–9588
Peakman M, Hill SJ (1994) Adenosine A2B-receptor-mediated cyclic AMP accumulation in
primary rat astrocytes. Br J Pharmacol 111:191–198
Pedata F, Dettori I, Coppi E, Melani A, Fusco I, Corradetti R, Maria A (2016) Neuropharmacology
purinergic signalling in brain ischemia. Neuropharmacology 104:105–130. https://doi.org/10.
1016/j.neuropharm.2015.11.007
Peleli M, Fredholm B, Sobrevia L, Carlström M (2017) Pharmacological targeting of adenosine
receptor signaling. Mol Aspects Med 55:4–8. https://doi.org/10.1016/j.mam.2016.12.002
Phosri S, Arieyawong A, Bunrukchai K (2017) Stimulation of adenosine A 2B receptor inhibits
endothelin-1-induced cardiac fibroblast proliferation and α-smooth muscle actin synthesis
through the cAMP/Epac/PI3K/Akt-signaling pathway. Front Pharmacol 8:1–15
Phosri S, Bunrukchai K, Parichatikanond W, Sato VH, Mangmool S (2018) Epac is required for
exogenous and endogenous stimulation of adenosine A 2B receptor for inhibition of angiotensin
II-induced collagen synthesis and myofibroblast differentiation. Purinergic Signal 14:141–156
Pinna A, Serra M, Morelli M, Simola N (2018) Role of adenosine - A2A receptors in motor control:
relevance to Parkinson’s disease and dyskinesia. J Neural Transm 125:1273–1286. https://doi.
org/10.1007/s00702-018-1848-6
Pleli T, Mondorf A, Ferreiros N, Thomas D, Dvorak K, Biondi RM, Heringdorf DMZ, Zeuzem S,
Geisslinger G, Zimmermann H, Waidmann O, Piiper A (2018) Activation of adenylyl cyclase
causes stimulation of adenosine receptors. Cell Physiol Biochem 45:2516–2528
Ponnoth DS, Nadeem A, Tilley S, Mustafa SJ (2010) Involvement of A 1 adenosine receptors in
altered vascular responses and inflammation in an allergic mouse model of asthma. Am J
Physiol Heart Circ Physiol 299:H81–H87
Preti D, Baraldi PG, Moorman AR, Borea PA, Varani K (2015) History and perspectives of A2A
adenosine receptor antagonists as potential therapeutic agents. Med Res Rev 35:790–848
Prystowsky EN, Niazi I, Curtis AB, Wilber DJ, Bahnson T, Ellenbogen K, Dhala A, Bloomfield
DM, Gold M, Kadish A, Fogel RI, Gonzalez MD, Belardinelli L, Shreeniwas R, Wolff AA
(2003) Termination of paroxysmal supraventricular tachycardia by tecadenoson (CVT-510), a
novel A 1-adenosine receptor agonist. J Am Coll Cardiol 42:1098–1102. https://doi.org/10.
1016/S0735-1097(03)00987-2
Rabadi MM, Lee HT (2015) Adenosine receptors and renal ischaemia reperfusion injury. Acta
Physiol 213:222–231
Raines J, Shakowski C, Page R, Quaife R (2019) Regadenoson associated side effect reversal:
safety and efficacy of IV theophylline. J Nucl Med 60:307
Romagnoli R, Baraldi PG, Moorman AR, Borea PA, Varani K (2015) Current status of A1
adenosine receptor allosteric enhancers. Future Med Chem 7:1247–1259
Rosenberger P, Schwab JM, Mirakaj V, Masekowsky E, Mager A, Morote-garcia JC, Unertl K,
Eltzschig HK (2009) Hypoxia-inducible factor–dependent induction of netrin-1 dampens
inflammation caused by hypoxia. Nat Immunol 10:195–202
Roshan MHK, Tambo A, Pace NP (2016) Potential role of caffeine in the treatment of Parkinson’s
disease. Open Neurol J 10:42–58
Ruiz MDL, Lim Y, Zheng J (2014) Adenosine A2A receptor as a drug discovery target. J Med
Chem 57:3623–3650
Ryzhov S, Zaynagetdinov R, Goldstein AE, Novitskiy SV, Dikov MM, Michael R, Biaggioni I,
Feoktistov I, Ryzhov S, Zaynagetdinov R, Goldstein AE, Novitskiy SV (2008) Effect of A2B
adenosine receptor gene ablation on proinflammatory adenosine signaling in mast cells. J
Immunol 180:7212–7220
Sachdeva S, Gupta M (2013) Adenosine and its receptors as therapeutic targets: an overview. Saudi
Pharm J 21:245–253. https://doi.org/10.1016/j.jsps.2012.05.011
Saki M, Yamada K, Koshimura E (2013) In vitro pharmacological profile of the A2A receptor
antagonist istradefylline. Naunyn Schmiedebergs Arch Pharmacol 386:963–972
10 Pharmacology of Adenosine Receptors 357
Samanta PN, Kar S, Leszczynski J (2019) Recent advances of in-silico modeling of potent
antagonists for the adenosine receptors. Curr Pharm Des 25:750–773
Sassi Y, Laggerbauer B, Sassi Y, Ahles A, Truong DJ, Baqi Y, Lee S, Husse B, Hulot J (2014)
Cardiac myocyte–secreted cAMP exerts paracrine action via adenosine receptor activation find
the latest version: cardiac myocyte–secreted cAMP exerts paracrine action via adenosine
receptor activation. J Clin Invest 124:5385–5397
Sawynok J (2016) Adenosine receptor targets for pain. Neuroscience 338:1–18. https://doi.org/10.
1016/j.neuroscience.2015.10.031
Schulte G, Fredholm BB (2000) Human adenosine A1, A2A, A2B, and A3 receptors expressed in
Chinese hamster ovary cells all mediate the phosphorylation of extracellular-regulated kinase
1/2. Mol Pharmacol 58:477–482
Schulte G, Fredholm BB (2003) Signalling from adenosine receptors to mitogen-activated protein
kinases. Cell Signal 15:813–827
Shah SJ, Voors AA, McMurray JJV, Kitzman D, Viethen T, Wirtz AB, Huang E, Pap AF, Solomon
SD (2019) Effect of neladenoson bialanate on exercise capacity among patients with heart
failure with preserved ejection fraction a randomized clinical trial. JAMA 321:2101–2112
Shaik K, Muttaleb A, Jaber Y, Kachler S, Klotz KN (2019) 7-Amino-2-aryl/hetero-aryl-5-oxo-5,8-
dihydro[1,2,4]triazolo[1,5-a] pyridine-6-carbonitriles: synthesis and adenosine receptor binding
studies. Chem Biol Drug Des 94:1568–1573
Sharma AK, Lapar DJ, Stone ML, Zhao Y, Mehta CK, Kron IL, Laubach VE (2016) NOX2
activation of natural killer T cells is blocked by the adenosine A2A receptor to inhibit lung
ischemia – reperfusion injury. Am J Respir Crit Care Med 193:988–999
Sitaraman SV, Wang L, Wong M, Bruewer M, Hobert M, Yun C, Merlin D, Madara JL (2002) The
adenosine 2b receptor is recruited to the plasma membrane and associates with E3KARP and
Ezrin upon agonist stimulation. J Biol Chem 277:33188–33195
Soares AS, Costa VM, Diniz C (2014) The combination of Cl-IB -MECA with paclitaxel: a new
anti-metastatic therapeutic strategy for melanoma. Cancer Chemother Pharmacol 74:847–860
Soni H, Peixoto-neves D, Buddington RK, Adebiyi XA (2017) Adenosine A 1 receptor-operated
calcium entry in renal afferent arterioles is dependent on postnatal maturation of TRPC3
channels. Am J Physiol Renal Physiol 313(313):1216–1222
Soudijn W, van Wijngaarden I, Ijzerman AP (2006) Allosteric modulation of G-protein–coupled
receptors. In: Bowery NG (ed). Allosteric receptor modulation in drug targeting. pp. 179–206
Stein E, Zou Y, Poo M, Tessier-Lavigne M (2001) Binding of DCC by netrin-1 to mediate axon
guidance independent of adenosine A2B receptor activation. Science (80-) 291:1976–1983
Stemmer SM, Madi L, Castel D, Ochaion A, Cohen S (2008) The A3 adenosine receptor agonist
CF102 induces apoptosis of hepatocellular carcinoma via de-regulation of the Wnt and NF- κ B
signal transduction pathways. Int J Oncol 33:287–295
Stemmer SM, Benjaminov O, Medalia G, Ciuraru NB, Silverman MH, Bar-Yehuda S, Fishman S,
Harpaz Z, Farbstein M, Cohen S, Patoka R, Singer B, Kerns WD, Fishman P (2013) CF102 for
the treatment of hepatocellular carcinoma: a phase I/II, open-label, dose-escalation study.
Oncologist 18:25–26
Stemmer SM, Manojlovic NS, Marinca MV, Petrov P, Cherciu N, Ganea D, Ciuleanu T-E, Puscas
IA, Beg MS, Purcell WT, Croitoru A-E, Ilieva RN, Natošević S, Nita AL, Kalev DN, Harpaz Z,
Farbstein M, Silverman MH, Fishman P, Llovet JM (2019) A phase II, randomized, double-
blind, placebo-controlled trial evaluating efficacy and safety of namodenoson (CF102), an A3
adenosine receptor agonist (A3AR), as a second-line treatment in patients with Child-Pugh B
(CPB) advanced hepatocellular carcinoma. J Clin Oncol 37:2503–2503
Stenberg DAG, Litonius E, Halldner L, Johansson RN, Fredholm BB (2003) Sleep and its
homeostatic regulation in mice lacking the adenosine A 1 receptor. J. Sleep Res 12:283–290
Stoilov RM, Licheva RN, Mihaylova MK, Reitblat T, Dimitrov EA, Shimbova KM, Bhatia G,
Pispati A, Balbir AG, Bagaria B, Oparanov BA, Fishman S, Harpaz Z, Farbstein M, Cohen S,
Bristol D, Silverman MH, Fishman P (2014) Therapeutic effect of oral CF101 in patients with
rheumatoid arthritis: a randomized, double-blind, placebo-controlled phase II study. Immunome
Res 11:1–6
358 P. K. Deb et al.
Sun Y, Huang P (2016) Adenosine A2B receptor: from cell biology to human diseases. Front Chem
4:1–11
Sun D, Samuelson LC, Yang T, Huang Y, Paliege A, Saunders T, Briggs J, Schnermann J (2001)
Mediation of tubuloglomerular feedback by adenosine: evidence from mice lacking adenosine
1 receptors. PNAS 98:9983–9988
Sun C, Schnermann J, Michael R, Sun C, Young HW, Molina JG, Volmer JB (2005) A protective
role for the A 1 adenosine receptor in adenosine-dependent pulmonary injury. J Clin Invest
115:35–43
Sun Y, Duan Y, Eisenstein AS, Hu W, Quintana A, Lam WK, Wang Y, Wu Z, Ravid K, Huang P
(2012) A novel mechanism of control of NF k B activation and inflammation involving A2B
adenosine receptors. J Cell Sci 125:4507–4517
Szentmiklósi AJ, Galajda Z, Cseppent Á, Hegyi B, Nánási PP (2015) The janus face of adenosine:
antiarrhythmic and proarrhythmic actions. Curr Pharm Des 21:965–976
Thomas GS, Cullom SJ, Kitt TM, Feaheny KM, Ananthasubramaniam K, Gropler RJ, Jain D,
Thompson RC (2017) The EXERRT trial: ‘“EXErcise to Regadenoson in Recovery Trial”’: a
phase 3b, open-label, parallel group, randomized, multicenter study to assess regadenoson
administration following an inadequate exercise stress test as compared to regadenoson with.
J Nucl Cardiol 24:788–802
Thompson C (2008) FDA approves pharmacologic stress agent. Am J Heal Pharm 65:890
Vallon V, Mu B (2006) Adenosine and kidney function. Physiol Rev 86:901–940
Varani K, Vincenzi F, Merighi S, Gessi S, Borea PA (2017) Biochemical and pharmacological role
of A 1 adenosine receptors and their modulation as novel therapeutic strategy. Adv Exp Med
Biol 1051:193–232
Vij A, Golzar Y, Doukky R (2018) Regadenoson use in chronic kidney disease and end-stage renal
disease: a focused review. J Nucl Cardiol 25:137–149
Vincenzi F, Targa M, Corciulo C, Gessi S, Merighi S, Setti S, Cadossi R, Borea PA, Varani K
(2012) The anti-tumor effect of A 3 adenosine receptors is potentiated by pulsed electromag-
netic fields in cultured neural cancer cells. PLoS One 7:e39317
Voelker R (2019) Add-on drug approved for “off” episodes of Parkinson disease. JAMA
322:1246–1246
Voors AA, Düngen H, Senni M, Agostoni P, Ponikowski P, Bax JJ, Voors PAA (2017) Safety and
tolerability of neladenoson bialanate, a novel oral partial adenosine A1 receptor agonist, in
patients with chronic heart failure. J Clin Pharmacol 57:440–451
Voors AA, Shah SJ, Bax JJ, Butler J, Gheorghiade M, Hernandez AF, Kitzman DW, McMurray
JJV, Wirtz AB, Lanius V, van der Laan M, Solomon SD (2018) Rationale and design of the
phase 2b clinical trials to study the effects of the partial adenosine A1-receptor agonist
neladenoson bialanate in patients with chronic heart failure with reduced (PANTHEON) and
preserved (PANACHE) ejection fraction. Eur J Heart Fail 20:1601–1610
Wang J, Huxley VH (2006) Adenosine A2A receptor modulation of juvenile female rat skeletal
muscle microvessel permeability. Am J Physiol Heart Circ Physiol 291:H3094–H3105
Wang L, Kolachala V, Walia B, Balasubramanian S, Hall RA, Merlin D, Sitaraman SV,
Kolachala V, Walia B, Bala S, Hall RA, Merlin D, Shanthi V (2004) Agonist-induced polarized
trafficking and surface expression of the adenosine 2b receptor in intestinal epithelial cells: role
of SNARE proteins. Am J Physiol Gastrointest Liver Physiol 287:1100–1107
Willingham SB, Ho PY, Hotson A, Hill C, Piccione EC, Hsieh J, Liu L, Buggy JJ, McCaffery I,
Miller RA (2018) A2AR antagonism with CPI-444 Induces antitumor responses and augments
efficacy to anti–PD-L1 and anti–CTLA-4 in preclinical models. Cancer Immunol Res 6:1–34
Wu LIN, Belardinelli L, Zablocki JA, Palle V, Shryock JC, Belardinelli L, Zablocki JA, Shryock JC
(2001) A partial agonist of the A1-adenosine receptor selectively slows AV conduction in
guinea pig hearts. Am J Physiol Heart Circ Physiol 280:334–343
Yaar R, Jones MR, Chen J, Ravid K (2005) Animal models for the study of adenosine receptor
function. J Cell Physiol 202:9–20
10 Pharmacology of Adenosine Receptors 359
Yang D, Wagner DD, Ravid K, Yang D, Zhang Y, Nguyen HG, Koupenova M, Chauhan AK,
Makitalo M, Schreiber BM, Gavras H, Wagner DD, Ravid K (2006) The A2B adenosine
receptor protects against inflammation and excessive vascular adhesion. J Clin Invest
116:1913–1923
Yang X, Xin W, Yang X-m, Kuno A, Rich TC, Cohen MV, Downey JM (2011) A2B adenosine
receptors inhibit superoxide production from mitochondrial complex I in rabbit cardiomyocytes
via a mechanism sensitive to Pertussis toxin. Br J Pharmacol 163:995–1006
Zhou J, Zimmermann K, Krieg T, Soltow M, Pavlovic D, Cerny V, Lehmann C (2015) Adenosine
receptor activation improves microcirculation in experimental intestinal ischemia/reperfusion.
Clin Hemorheol Microcirc 59:257–265
Zimmermann H (2000) Extracellular metabolism of ATP and other nucleotides. Naunyn
Schmiedebergs Arch Pharmacol 362:299–300
Pharmacology of Angiotensin and Its
Receptors 11
Satyajeet Biswal, Rajat Ghosh, and Pratap Chandra Acharya
Abstract
angiotensin as drug target and the use of ARBs for the pharmacotherapeutic
intervention of hypertension.
Keywords
Abbreviations
11.1 Introduction
11.1.1 History
11.1.2 Discovery
The pressor principle ‘renin’ was extracted from the kidney by Tigerstedt and
Bergman in 1897 and provided the first insight into the regulation of blood pressure.
This revolutionary work further directed the discovery of reno-vascular hypertension
in mammals (Goldblatt et al. 1934). When Goldblatt isolated the vasoconstrictor
substance renin from the renal venous blood of a hypertensive dog, nobody believed
it till 1940 (Braun-Menendez et al. 1940). A similar discovery was made simulta-
neously and independently by Page and Helmer in 1940, which showed that the
so-called renin activator, later proved to be angiotensinogen, can be isolated from the
intact animal after injecting it with renin. In Argentina, the pressor substance was
known to be ‘hypertension’ whereas in the United States of America it was called
angiotonin and was later shown to be an octapeptide (Skeggs Jr. et al. 1956; Bumpus
et al. 1957; Elliott and Peart 1956). There were differences between laboratories
concerning the nomenclature, but it was later established that both hypertensin and
angiotonin were the same octapeptide, and the hybrid term ‘angiotensin’ was
unanimously accepted by Braun-Mene’ndez and Page.
Components of angiotensin II-forming cascade such as angiotensinogen,
angiotensin-converting enzyme (ACE), and angiotensins I, II and III were further
characterized. In 1987, a committee was formed comprising members from the
‘International Society for Hypertension’, ‘The American Heart Association’ and
the ‘World Health Organization’ which proposed to abbreviate angiotensin to Ang
using the decapeptide angiotensin I as the reference for numbering the amino acid
sequence of all angiotensin peptides (Dzau et al. 1987). Ang II regulates vascular
resistance and blood volume and has been found in tissues such as kidney, adrenals,
brain, sympathetic nervous system, pituitary gland and vascular smooth muscle. Ang
II is also suggested to function as a paracrine and autocrine hormone in the regula-
tion of cellular growth, proliferation and extracellular matrix formation (Dzau and
Gibbons 1987; Grady et al. 1991; Weber et al. 1995b; Weber et al. 1995c).
Metabolites of angiotensin such as angiotensin 2–8 (Ang III), angiotensin 1–7 or
angiotensin 3–8 (Ang IV) have also been shown to exhibit biological activities
(Peach 1977; Schiavone et al. 1990; Chappell et al. 1991; Iyer et al. 1998; Wright
and Harding 1995).
At the end of the 1980s, it was demonstrated that at least two receptor types exist
for Ang II in many tissues. The conventional peptide analogue such as saralasin and
nonpeptide antagonist such as losartan were found to have a high affinity for both the
receptors but without any selectivity (Chiu et al. 1989; Whitebread et al. 1989; Speth
and Kim 1990). The initial nomenclature of the receptor subtypes was confusing, for
example the receptor sensitive to losartan was called 1, B or a, whereas the receptor
with no affinity for losartan was termed 2, A or b. The ‘High Blood Pressure
Research Council’ in 1990 and the ‘International Union of Pharmacology Commit-
tee on Receptor Nomenclature and Drug Classification’ (NC-IUPHAR) appointed a
subcommittee to address the problem, and a classification was proposed in 1991 and
updated in 1995 (Bumpus et al. 1991; De Gasparo et al. 1995). The details of the
receptor subtype classification are described in Sect. 11.3.
366 S. Biswal et al.
Liver
Secretes Angiotensinogen
Angiotensinogen
Renin
Converts Angiotensinogen
to Angiotensin I
Kidney
Releases Renin
Angiotensin I
ACE
Converts Angiotensin I
Lungs
to Angiotensin II Releases Angiotensin
Converting Enzyme
Angiotensin II
(ACE)
Angiotensin II Receptors
Located in Adrenal glands, Vascular smooth muscle, the heart and the brain
Angiotensin stimulates Angiotensin stimulates Angiotensin stimulates Angiotensin stimulates Angiotensin stimulates
aldosterone secretion sodium & fluid retention in muscle hypertrophy & sympathetic outflow in the vasoconstriction in blood
in the adrenal gland. the kidney fibrosis in the heart brain vessels
11.2 Angiotensin
The enzyme renin and its greater precursor, prorenin, are produced by the zona
glomerulosa cells present in renal afferent arterioles of the kidney (Fig. 11.1). The
regulation of the renin–angiotensin–aldosterone system is dependent on several
enzymes with the end product being Ang II. The physiological action of Ang II is
further mediated by the interaction of the peptide with the specific cell surface
receptors namely AT1 and AT2 (Guthrie Jr. 1995).
During the circulation, Ang II contracts vascular smooth muscle by which elevation
of blood pressure happens due to the resistance developed in the arterioles. The renal
and mesenteric beds are known to be most sensitive to the Ang II action. Arteriolar
smooth muscle hypertrophy and increased peripheral resistance are also contributed
by Ang II by acting as a growth factor for blood vessels. This action is particularly
important in the case of vascular injury (Naftilan 1992).
11 Pharmacology of Angiotensin and Its Receptors 367
Ang II is known to play a major role in the fluid and electrolyte balance as well as
cardiovascular functions. However, the circulating Ang II can activate the
angiotensinergic sympatho-excitatory pathway in the brain leading to the develop-
ment and progression of hypertension. Evidences indicate that central sympathetic
nerve activity can trigger pathogenicity of essential hypertension. The sympatho-
adrenomedullary system is known to have an adaptive response to stress. However,
excessive or sustained stress can contribute to the state of hypertension (Mcdougall
et al. 2005; Esler 2009).
Ang II, the peptide hormone, sustains blood pressure and vascular volume
through several actions such as vasoconstriction, stimulation of aldosterone secre-
tion, increased renal tubular absorption of sodium, activation of the sympathetic
nervous system and increased cardiac contractility. However, these actions are
impaired in the pathophysiologic states of hypertension and congestive heart failure
(CHF). Increased release of renin and Ang II has been found in renal diseases like
renal artery stenosis, renin-secreting tumours or kidney injury leading to hyperten-
sion (Guthrie Jr. 1995).
Therefore, drugs such as ACE inhibitors and ARBs are able to antagonize the
actions of the renin–angiotensin axis leading to effective treatment of hypertension
and heart failure.
et al. 1991; Sasaki et al. 1991; Kambayashi et al. 1993; Mukoyama et al. 1993).
However, the biochemical and physiological functions of the AT2 receptor are still a
matter of intense research.
11.3.1.3 AT3 Receptor
The existence of AT3 receptor subtype has been reported with its characteristic
pharmacology. However, the distinct gene for this receptor in humans has not
been established to date (Chaki and Inagami 1992; Inagami et al. 1993).
11.3.1.4 AT4 Receptor
High-affinity binding sites of the Ang IV peptide were found to be concentrated
predominantly in the brain and to some extent in heart, kidney, adrenals and blood
vessels, which were termed as AT4 receptors in 1995 (Harding et al. 1992). The AT4
receptor does not bind the peptide analogues of Ang II as well as the non-peptide
inhibitors of AT1 and AT2 receptors such as losartan, CGD42112A and PD123177
(Karnik et al. 2015).
hypertensive rats (SHR), which cannot be done by the vasodilator drug hydralazine
(Antonaccio et al. 1979). This advanced research resulted in a number of in vitro
studies showing the positive effect of Ang II on cardiomyocytes and vascular smooth
muscle cells and was identified as a promising cardiovascular growth factor.
Single dose a day, negligible adverse effect, patient tolerance to side effect and most
importantly the lower cost make these sartans the first-line choice as antihyperten-
sive drugs. These ARBs have a good tolerance profile as compared to the ACE
inhibitors in both long term and short term due to which the patient compliance is
much higher than other antihypertensive drugs. In the initial phase, hypertension is
asymptomatic, and long-term treatment is necessary to control the blood pressure.
Patient compliance is very much essential, and there should not be any interaction
with food. Sartans come into this category and make oral administration very easy
(Guthrie Jr. 1995).
non-steroidal antiinflammatory drugs (NSAIDS) are not class specific, and clinically
it may interfere with the antihypertensive effect of ARBs.
The clinical status of various angiotensin-II antagonists or ARBs has been compiled
in Table 11.2 and described in the following section.
Azilsartan
Azilsartan (Fig. 11.2) is the latest Ang II receptor blocker, developed by Takeda
Pharmaceuticals in the brand name Edarbi, and has been approved by the United
States Food and Drug Administration (US FDA) on 25 February 2011. Azilsartan is
well accepted in the market because of its sustained blood pressure control, which
lasts for 24 h. It is a potent and highly selective Ang II receptor blocker having a
bioavailability of 60% and an elimination half-life of 11 h. The prodrug azilsartan
medoxomil gets hydrolysed to azilsartan very quickly. The dose range of 40 mg or
80 mg once daily shows promising results in systolic and diastolic blood pressure by
reducing 12–15 mm Hg and 7–8 mm Hg respectively. Till now azilsartan is
prescribed only for hypertension, and there are no sufficient human data supporting
the use of azilsartan for the improvement of cardiovascular outcomes.
Candesartan
In the year 1993, candesartan (Fig. 11.3) was first examined by Japanese
scientists, and they published the effectiveness of the compound as an angiotensin
receptor blocker (Mizuno et al. 1992; Ogihara et al. 1993). Candesartan is available
in the market as its prodrug candesartan cilexetil (cyclohexyl 1-hydroxyethyl car-
bonate), which is an ester and gets completely metabolized to its active molecule
candesartan. Candesartan has much lower bioavailability when compared with other
ARBs. It has an absolute bioavailability of 15–40% and an elimination half-life of
9 h. Candesartan can be given in a dose range of 4–16 mg/day for better therapeutic
effectiveness. Candesartan is proved to be lethal if it is taken by pregnant women
during the second or third trimester. Patients with renal artery stenosis are very prone
to face high risk because of the reduction in renal glomerular filtration rate.
Irbesartan
In the year 1990, irbesartan (Fig. 11.4) was patented for use in the treatment of
high blood pressure and heart failure.
Irbesartan is available in the market under the brand name Avapro. This drug is
chosen as an initial treatment for high blood pressure. The dose range of this drug is
150–300 mg. Bioavailability is 60–80% and has an elimination half-life of 11–15 h.
Losartan
Losartan (Fig. 11.5) is the first discovered Ang II receptor antagonist, which was
patented in the year 1986 and got approved by US FDA in 1995. Because of its
effectiveness and less adverse effect, it got placed on the WHO (World Health
Organization) List of Essential Medicines. The primary function of the drug is to
lower the blood pressure, but it shows promising results in the case of renal disease
reduction in patients with type-2 diabetes, hypertension and microalbuminuria or
11
Table 11.2 Pharmacological characteristics of the angiotensin II receptor antagonists available on the market
Drug name Trade name Prodrug Bioavailability % Dose recommended (mg/day) Half-life (h) Protein binding %
Azilsartan Edarbi Azilsartan medoxomil 60 40–80 11 99
Candesartan Atacand Candesartan cilexetil 42 4–16 9 >99
Irbesartan Avapro – 60–80 150–300 11–15 90
Losartan Cozaar – 25–35 50–100 1.5–2 99.7
Pharmacology of Angiotensin and Its Receptors
O HN
N N
O O
O N
N NH
N
N
N
O
O N
OH
11 Pharmacology of Angiotensin and Its Receptors 375
N
N N
NH
O O O
O
N O
HO
proteinuria. However, calcium channel blockers and thiazide diuretics are prescribed
to most patients, but patients who have intolerance to ACE inhibitors are prescribed
with losartan. The bioavailability of losartan is around 25–35% and the elimination
half-life is 1.5–2 h.
Olmesartan
The drug olmesartan (Fig. 11.6) got approved by the FDA in April 2002 for the
treatment of hypertension. Olmesartan medoxomil is a prodrug and gets completely
metabolized to its active metabolite olmesartan rapidly. The bioavailability of
olmesartan is around 26%, and it reaches its peak plasma concentration in 1 or
2 h. The oral dose of olmesartan is 20–40 mg once in a day. Sometimes a diuretic can
be given with olmesartan to the patient when blood pressure cannot be controlled by
only olmesartan.
Telmisartan
Telmisartan (Fig. 11.7) is a nonpeptide Ang II receptor antagonist, which got
FDA approval in the year 1998 for use in the treatment of hypertension.
After oral administration, the peak plasma level is obtained in 0.5–1 h. The
bioavailability of telmisartan is around 42–100% and protein binding is high at
more than 99.5%. The elimination half-life is 24 h. Due to the biliary secretion of the
O
N N HO
11.6 Conclusions
The physiological effect of the peptide hormone angiotensin was unknown till the
discovery of two major angiotensin receptors, AT1 and AT2. Research on the
physiological role of angiotensin has led to the finding that Ang II is the primary
culprit in the development of hypertension and related cardiac diseases. However,
Ang II is also known to maintain the fluid and electrolyte balance in the body.
Therefore, the effective treatment of hypertension and heart failure can be achieved
by blocking the angiotensin receptors using ARBs or angiotensin antagonists. The
ARBs have been proven to be a better therapeutic intervention for the management
of hypertension than the ACE inhibitor, due to its fewer adverse effects, although
they both share the same biochemical pathway for the regulation of blood pressure.
In the recent years, the ARBs, also known as ‘saratans’, have been the corner-
stone in the management of hypertension and heart failure. The sartans are typically
the AT1 receptor blockers without any effect on AT2 receptor. It has been more than
three decades since the identification of the AT2 receptor. However, the biochemical
and physiological functions of the AT2 receptor are yet to be established. It is still a
matter of debate whether the AT2 receptor is involved in ovulation, neural tissue
regeneration and wound healing process. Therefore, at this juncture when the role of
AT1 receptor has been fully understood, the exploration of physiological properties
of AT2 receptor is warranted. This could possibly lead to the development of
selective AT2 receptor modulators for the management of diseases other than of
cardiovascular system.
References
Antonaccio MJ, Rubin B, Horovitz ZP, Laffan RJ, Goldberg ME, High JP, Harris DN, Zaidi I
(1979) Effects of chronic treatment with captopril (sq 14, 225), an orally active inhibitor of
angiotensin i-converting enzyme, in spontaneously hypertensive rats. Jpn J Pharmacol
29:285–294
Benz J, Oshrain C, Henry D, Avery C, Chiang YT, Gatlin M (1997) Valsartan, a new angiotensin II
receptor antagonist: a double-blind study comparing the incidence of cough with lisinopril and
hydrochlorothiazide. J Clin Pharmacol 37:101–107
11 Pharmacology of Angiotensin and Its Receptors 377
Siebers MJ, Goodfriend TL (1986) Platelet angiotensin receptors in young and old humans. J
Gerontol 41:574–578
Simpson RU, Campanile CP, Goodfriend TL (1980) Specific inhibition of receptors for angiotensin
II and angiotensin III in adrenal glomerulosa. Biochem Pharmacol 29:927–933
Skeggs LT Jr, Lentz KE, Kahn JR, Shumway NP, Woods KR (1956) The amino acid sequence of
hypertensin. II. J Exp Med 104:193–197
Speth RC, Kim KH (1990) Discrimination of two angiotensin II receptor subtypes with a selective
agonist analogue of angiotensin II, p-aminophenylalanine6 angiotensin II. Biochem Biophys
Res Commun 169:997–1006
Speth RC, Rowe BP, Grove KL, Carter MR, Saylor D (1991) Sulfhydryl reducing agents distin-
guish two subtypes of angiotensin II receptors in the rat brain. Brain Res 548:1–8
Timmermans PB, Wong PC, Chiu AT, Herblin WF, Benfield P, Carini DJ, Lee RJ, Wexler RR,
Saye JA, Smith RD (1993) Angiotensin II receptors and angiotensin II receptor antagonists.
Pharmacol Rev 45:205–251
Tufro-Mcreddie A, Johns DW, Geary KM, Dagli H, Everett AD, Chevalier RL, Carey RM, Gomez
RA (1994) Angiotensin II type 1 receptor: role in renal growth and gene expression during
normal development. Am J Phys 266:F911–F918
Weber K, Sun Y, Cleutjens J (1995a) Structural remodeling of the myocardium postinfarction:
potential mechanisms and influence of therapy. Cardiol Rev 3:53–65
Weber KT, Sun Y, Campbell SE (1995b) Structural remodelling of the heart by fibrous tissue: role
of circulating hormones and locally produced peptides. Eur Heart J 16 Suppl N:12–18
Weber KT, Sun Y, Katwa LC, Cleutjens JP (1995c) Connective tissue: a metabolic entity? J Mol
Cell Cardiol 27:107–120
Whitebread S, Mele M, Kamber B, De Gasparo M (1989) Preliminary biochemical characterization
of two angiotensin II receptor subtypes. Biochem Biophys Res Commun 163:284–291
Wright JW, Harding JW (1995) Brain angiotensin receptor subtypes AT1, AT2, and AT4 and their
functions. Regul Pept 59:269–295
Pharmacology of Endogenous Opioids,
Opiates and Their Receptors 12
Mohammed Noorladeen Al–Qattan, Nirupam Das,
and Rati Kailash Prasad Tripathi
Abstract
The search for analgesia is the main drive for the discovery of opioid receptors
and their endogenous ligand peptides. Although opioid peptides are very similar
in their N-terminal sequence, they are classified into different types according to
their precursor proteins. The peptides activate opioid receptors by binding
to orthosteric-binding sites to mediate intracellular second messengers. Biased
signaling and allosteric modulation is a new approach to obtain receptor subtype
selectivity and separates the desirable from a myriad of unwanted pharmacologi-
cal effects. This chapter describes endogenous opioids and opiates with an
emphasis on structure, origin and processing, receptors, physiological roles, and
potential involvement in therapeutic interventions. Further, it also provides a brief
discussion on the effect of opioids on various ion channels and recent
developments of established and investigational opioid molecules.
Keywords
Opioids receptors · Endorphin · Enkephalin · Dynorphin · GPCR · Neurological
disorders · Ion channels
Abbreviations
ACTH Adrenocorticotropin
BACE1 Beta-site APP cleaving enzyme 1
M. N. Al–Qattan
Department of Pharmacy, Al–Noor University College, Mosul, Iraq
N. Das (*) · R. K. P. Tripathi
Department of Pharmaceutical Science, Sushruta School of Medical and Paramedical Sciences,
Assam University, Silchar, Assam, India
12.1 Introduction
The inscription of the ancient Sumerian clay tablet insinuates the use of the natural
extract of poppy plant (Papaver somniferum) as analgesia and post-surgical pain
(Norn et al. 2005; Brownstein 1993). The active ingredient of the extract is mor-
phine, which necessitates the presence of receptors for binding inside the human
body. The search for endogenous ligands acting on those opioid receptors (ORs)
started in the 1960s and succeeded in a few years by using radiolabeled ligands (Pert
and Snyder 1973). On the search for ligands for those ORs, several peptides were
isolated. The early peptides were enkephalins (Hughes et al. 1975) which showed
higher potency than morphine on opioid receptors (Kosterlitz and Hughes 1975).
Soon the untriakontapeptide (endorphin) was discovered (Li and Chung 1976), then
dynorphins (Cox et al. 1975) and endomorphins (Zadina et al. 1997).
Although opioid peptides (OPs) share almost similar N-terminal amino acid
sequence, their precursor proteins are different. The processing of precursor proteins
differs from one tissue to another as well as variation of the half-life of opioid
peptides. Some endogenous opioid peptides like endomorphins act as a selective
agonist for μ-opioid receptor (MOR) and thus have a good potential for clinical use.
However, the low oral bioavailability of endomorphins restricts its use unless given
as glycosylated derivatives (Varamini et al. 2012). Currently, three types of opioid
receptors are known, namely Mu, Delta, and Kappa opioid receptors (MORs, DORs
and KORs, respectively). The receptors can be activated by orthosteric ligands and
cooperated with allosteric ligands to produce modulation of orthosteric affinity/
efficacy as well as biasing the intracellular signaling.
Endogenous opioid peptides are classified into three categories, namely endorphins,
enkephalins, and dynorphins. The peptides are endogenously produced by
peptidases digestion of larger precursor proteins, namely proopiomelanocortin,
12 Pharmacology of Endogenous Opioids, Opiates and Their Receptors 383
POMC
± oligosaccharide
± ±
+
ACTH biosynthec intermediate β-LPH
+ +
± ±
Fig. 12.1 The processing of POMC (Retrieved from Mains and Eipper 1981)
Table 12.1 Opioid peptides and their precursor proteins and receptor selectivity (Luca et al. 2007; Coward et al. 1998)
Endogenous peptide Amino acid sequence Receptor affinity Precursor
β-Endorphin YGGFMTSEKSQTPLVTLFKNAIIKNAYKKGE δ¼μ Proopiomelanocortin
[Met]-enkephalin YGGFM (YGGFMRF, YGGFMRGL) δ >> μ Proenkephalin
[Leu]-enkephalin YGGFL
Metorphinamide YGGFMRRV-NH2
Dynorphin A YGGFLRRIRPKLKWDNQ κ >> δ ¼ μ Prodynorphin
Dynorphin A(1-8) YGGFLRRI
Dynorphin B YGGFLRRQFKVVT
α-neoendorphin YGGFLRKYPK
YGGFLRKYP
β-neoendorphin
Nociceptin FGGFTGARKSARKLANQ *ORL Pronociceptin
Endomorphin-1 YPWF-NH2 μ Unknown
Endomorphin-2 YPFF-NH2
ORL means orphan opioid-receptor-like
Pharmacology of Endogenous Opioids, Opiates and Their Receptors
385
386 M. N. Al–Qattan et al.
Endomorphins (1 and 2) are reported to be the only discovered opioid peptide that
are selective for MOR (Zadina et al. 1997). The two peptides differ from other opioid
peptides in having NH2-Tyr-Pro-Trp(Phe)-Phe-CO-NH2, that is, having
carboxamide (aminocarbonyl) terminal. Some studies showed that endomorphin-1
produces higher analgesia and without reward effect compared to morphine (Wilson
et al. 2000). Although the reward effect is known to be mediated by binding MOR, it
is limited to a specific conformational set of receptors; thus it is ligand-dependent
property. However, the potential of respiratory depression, urinary retention, toler-
ance, addiction and cardiac side effects and the low intrinsic bioavailability restrict
the clinical use of endomorphins (Gu et al. 2017). Glycosylation through succinamic
acid linker at the N-terminal enhances metabolic stability and membrane permeabil-
ity of endomorphins while maintaining potency and efficacy (Varamini et al. 2012).
Nociceptin is structurally related to dynorphin; however, it binds to opioid
receptor-like (ORL) while having no affinity for other ORs. Similar to dynorphin,
nociceptin antagonizes the analgesic effect of other opioid ligands (Mika et al.
2011). Activation of ORL inhibits adenylyl cyclase and Ca+2 channels
while activating K+ channels like opioid receptors (Calo et al. 2000). However, the
pharmacological behavior observed upon activation by nociceptin produces potent
anti-analgesic action supra-spinally and analgesic action spinally (Mogil and
Pasternak 2001).
Fig. 12.2 Crystal structure of active conformation of Mu opioid receptors (PDB ID: 6DDE) with
missing H8 being added from PDB 5C1M and the position within cell membrane is optimized using
OPM database
without the concomitant dysphoria and sedation usually associated with this receptor
(Brust et al. 2016). Several biased agonists at KOR were successfully developed, and
some are being used clinically (Mores et al. 2019; Brust et al. 2016). Despite the
previous successes, there are unresolved experimental limitations of measuring
differences in biased factor among various ligands in addition to the implications
of cellular environments (Mores et al. 2019; Ho et al. 2018). Although beneficial
results observed by using biased agonists to stabilize receptor conformational
ensembles that said to activate intracellular Gαi over β-arrestin-2 recruitment
(Ranjan et al. 2017), such simplification of the story might not be precise (Conibear
and Kelly 2019; Bermudez et al. 2019).
Like other GPCRs, the allosteric sites of ORs are therapeutically promising to get
biased signaling (Livingston and Traynor 2018; Livingston et al. 2018). Moreover,
allosteric ligands can enhance efficacy/affinity for endogenous opioid peptides as
well as provide selectivity toward particular OR type, which usually is difficult to
achieve using orthosteric ligands (Livingston et al. 2018). Interestingly, some
endogenous compounds act as PAM for ORs, for example, oxytocin which is a
peptide hormone principally involved in labor and lactation act as PAM for
orthosteric endomorphin-1, β-endorphin, and morphine by enhancing efficacy and
not affinity toward MOR (Meguro et al. 2018). The tuber of the species Aconitum is
traditionally used in Japan to relieve pain, currently shown to have ignavine, that has
selectivity for MOR over KOR and acts as PAM for endomorphin-1 and morphine
(Ohbuchi et al. 2016). Biased signaling can also be produced using allosteric
modulators. The BMS-986187, a synthetic compound discovered by high-
throughput screening (HTS), can function as PAM selectively for DOR (Burford
et al. 2015) and produce biased signaling toward G-protein over β-arrestin-2, which
is elicited by lower receptor internalization (Stanczyk et al. 2019).
HO
H2N NH
O H O O
O H
N
N CH3 N N NH2
H
H
O
HO
1 2
H2N
O O
O H H
O O H3 C O
H O
N
N NH2 CH3
N O
H
O
CH3
O O
3 CH3
4
12 Pharmacology of Endogenous Opioids, Opiates and Their Receptors 395
SCH3
O
H
HOOC N
H H
O HN
H O HOOC N
N O
H2N H H
N
H 6
H
5
HN NH2 HN NH2
OH
NH NH
O O O
H H
N N N
N N N
O NH H H H
O O O NH
O O
HN
NH HN
O
O NH HN NH2
O
7 HN
NH2
H2N
O
OH
OH NH2
The dynorphins were profoundly expressed during the aging process and AD and
have been reported to stimulate the KOR and thought to induce stress-related
memory impairments. Additionally, they also affect glutamate neurotransmission
and perturb their function of synaptic plasticity essential for memory (Ménard et al.
2013).
The rate-determining step in the production of Aβ is the proteolytic breakdown of
Amyloid Precursor Protein (APP) by β-secretase (BACE1). The activation of DOR
396 M. N. Al–Qattan et al.
by an agonist has been shown to increase the secretase activity probably through the
post-translational mechanism and causing an increased formation of Aβ
(Sarajarvi et al. 2015). The cascade of reaction makes DOR and BACE1 as prospec-
tive targets in the design of DOR antagonist (Zhao et al. 2015) and BACE1 inhibitors
(Coimbra et al. 2018) for ameliorating the neurodegeneration underlying AD. The
hypermethylation of opioid receptor δ 1 (OPRD1) promoter was also linked with the
risk of AD (Ji et al. 2017). Subsequent studies suggest that in addition to OPRD1,
elevated methylation of opioid receptor κ1 and opioid receptor μ1 genes are also
involved in the progression of AD. Therefore, the genes of the opioid receptors could
serve as potential biomarkers for AD diagnosis (Xu et al. 2018).
12.4.2 Schizophrenia
Schizophrenia is a multifaceted, diverse mental disorder that affects the behavior and
cognitive function and has genetic or environmental predisposition, or both.
Antipsychotics, along with psychological therapies, are the primary line of manage-
ment available to alleviate the disorder. In recent years, much research to gain insight
into the pathophysiology of schizophrenia has been undertaken (Owen et al. 2016;
Patel and Shulman 2015) and identification of novel targets is under investigation
(Gill et al. 2018; Yang and Tsai 2017). The cardinal features of schizophrenia are
negative symptoms (reduced enthusiasm and withdrawal from society), cognitive
symptoms (disruption of attentiveness and dementia), and positive symptoms
(hallucinations accompanied by delusions). KOR agonists could elicit these specific
symptoms and drugs blocking this receptor might result in a fruitful therapeutic
outcome. Antipsychotics are capable of effectively combating the positive
symptoms; however, presently, efficient therapeutic managements for controlling
the symptoms (negative or cognitive) of schizophrenia are not available. The
potentiality of a pan-opioid antagonist either naloxone (8) or naltrexone (9) could
make KOR a promising target option for overall treatment benefits in schizophrenia
(Clark and Abi-Dargham 2019; Shekhar 2019). Alongside, contemporary research
on the discovery of novel KOR antagonist by Guerrero et al. has shown encouraging
results. The promising drug candidate BTRX-335140/CYM-53093 (10) exhibited
potent (IC50-0.8 nM) and selective KOR antagonistic activity with a favorable
pharmacokinetic profile. Currently, the compound is undergoing phase I clinical
trials for the possible treatment of various psychiatric disorders (Guerrero et al.
2019). A recent double-blind phase II study found that a combination of olanzapine
and fixed dose of MOR antagonist samidorphan (11) demonstrated clinically and
statistically significant reduction of weight gain and adverse metabolic effect of
olanzapine without compromising the antipsychotic efficacy of olanzapine. The
combination was considerably tolerated and comparable to that of olanzapine-
placebo in terms of safety (Chaudhary et al. 2019; Martin et al. 2019).
12 Pharmacology of Endogenous Opioids, Opiates and Their Receptors 397
HO HO
O OH O OH
N N
O O
8 9
O
O N H2N
N HO
N N O N
OH
F
N O
10 H 11
Post-traumatic stress disorder (PTSD) is a psychiatric disorder that may arise after a
single encounter or exposures to life-threatening chronic events. PTSD deteriorates
physical health and is mostly accompanied by cardiorespiratory, musculoskeletal,
gastrointestinal, immunological, endocrine, and metabolic problems. It is also
associated with psychiatric comorbidity and an increased suicidal tendency (Bisson
et al. 2015; Yehuda et al. 2015). Existing approaches in the treatment of PTSD
involve cognitive behavioral therapy and the use of anxiolytic/antidepressant agents
to ameliorate the symptoms (Shalev et al. 2017). Interestingly, studies suggest that
endogenous opioid peptides exhibited a placebo effect in PTSD, and the mood-
enhancing effects of the peptides may be initiated by exercise and light therapy to
relieve the stress. It is suggested that the interaction between dopaminergic pathways
and the endogenous opioids may be responsible for the placebo effect (Sher 2004),
although it would not be a stand-alone option and may be concomitantly utilized
along with standard drugs. As discussed earlier, the dynorphin mediates its action via
KOR and the dynorphin/KOR interrelationship is associated in several brain
disorders (De Lanerolle et al. 1997; Mathieu-Kia et al. 2001; Mello and Negus
2006). Various works of the literature suggest that in PTSD, there is a substantial
expression of the KOR and mediate the symptoms of anxiety. Therefore, in line of
the evidence, targeting the KOR might be a viable option in the management of
PTSD (Bailey et al. 2013). The opioid analgesics prescribed in PTSD often result in
comorbidity between PTSD and OUD, and they are frequently considered as two
sides of the same coin (Elman and Borsook 2019; Hassan et al. 2017).
Centrally acting competitive MOR antagonist opioid receptor antagonists such as
naloxone is the ideal choice in emergencies related to opioid overdose. On the
contrary, naltrexone, which mediates its action via KOR antagonism, is employed
mainly in OUD and AUD for maintaining abstinence by decreasing the cravings
398 M. N. Al–Qattan et al.
(Theriot et al. 2019). However, both opioid agonist and antagonist are utilized in
substance abuse therapies to combat the withdrawal syndromes and for the inhibition
of return usage. Agonists such as morphine and methadone (12), partial agonist
buprenorphine (13) and opioid antagonist such as extended-release injectable nal-
trexone are recommended for overall treatment and tackling the relapsing of OUD.
The mechanism by which opioid antagonist maintain abstinence in OUD and AUD
is by reducing the mesolimbic dopaminergic neurotransmission (McCarty et al.
2018; Williams et al. 2008).
HO
N
O
O
O
OH
H
12 13
locally microinjected in the globus pallidus (GP), increased akinesia was observed,
and vice versa, when injected in the substantia nigra pars reticulate. Adverse effects
such as convulsions may restrict the use of DOR agonist in Parkinsonism. The
convulsion was avoided by a synergistic combination of a DOR agonist, SNC-80,
(15) and J-113397, an N/OFQ antagonist (Mabrouk et al. 2009; Mabrouk et al.
2014). Conversely, UF-512 showed encouraging activities as anxiolytic/antidepres-
sant and treatment against chronic and neuropathic pain (Polo et al. 2019; Vergura
et al. 2008). Recently, a new mixed DOR agonist/MOR antagonist, DPI-289 (16), in
combination with levodopa elicited improved activity without increasing dyskinesia,
and it was superior when compared to high dose levodopa (Johnston et al. 2018).
HO
H2N
O O
O N N
HN O N
OH
N N
HN
O 15
14
N
H
OH
16
400 M. N. Al–Qattan et al.
Mental illness such as bipolar disorder (BD) and major depressive disorder (MDD)
are recalcitrant to treat due to the chronic nature and due to inter-individual variation
(Jeon et al. 2016). Studies found that opioid analgesic demonstrated potent mood-
elevating effect on patients with bipolar disorder and involve positive interaction
between the opioid and the dopaminergic systems (Schaffer et al. 2007). Preclinical
evaluations revealed that the dynorphin system is related to mood, motor, cognitive,
and endocrine functionality and subjects with MDD and BD showed a decreased
level of prodynorphin mRNA expression (Hurd 2002). Besides, a novel KOR
antagonist MCL-144B (17) displayed antidepressant activity in the forced swim
test (Reindl et al. 2008). Berrocoso et al. reported that a combination of a selective
serotonin reuptake inhibitor (SSRI) with a weak MOR agonist, (+)-tramadol (18),
produced better antidepressant activity than SSRI alone (Berrocoso and Mico 2009).
As previously discussed, UF-512, a DOR agonist, showed anxiolytic/antidepressant
properties (Polo et al. 2019). However, a combination of opioid-based samidorphan
(MOR antagonist) and buprenorphine (ALKS 5461) in phase III trials was recently
rejected by USFDA because of inadequate data to prove its effectiveness.
N
N
O H
O
H
O CH2)8 O 2HCl
17
OH
18
The body of human beings normally generates substances similar to opiates and
utilizes them as neuromodulators. These opiate-like substances comprise of
β-endorphins, dynorphins, and enkephalins, and are frequently conjointly called as
opioid peptides or endogenous opioids (Corder et al. 2018; Li et al. 2012; Pathan and
Williams 2012). Opioid peptides are implicated in the regulation and/or control of
various body functions such as tolerance and drug dependency, stress and pain,
12 Pharmacology of Endogenous Opioids, Opiates and Their Receptors 401
The entry of Ca2+ ions through the voltage-gated Ca2+ channels (VGCCs) results in
depolarization of nerve terminals that further causes the discharge of
neurotransmitters from the nerve cells. Three types of voltage-gated Ca2+ channels
are reported, namely, T-type channels which are capable of showing small conduc-
tance, N-type channels which are inept to demonstrate intermediate conductance,
and L-type channels which illustrate large conductance. Opioids act through the
inhibition of N-type voltage-gated Ca2+ channels and reduce the passage of Ca2+
ions inside the cell, thereby inhibiting the release of neurotransmitters (Zamponi
et al. 2015; Seseña et al. 2014; Catterall et al. 2013; Zamponi and Currie 2013).
However, this action of opioids exclusively is not accountable for the total cumula-
tive effect of opioids on neurotransmitter release (Chieng and Bekkers 2001).
The euphoric effect of opioids may be due to another mechanism in which the
GABA inhibitory interneurons of the ventral tegmental area (VTA) are involved
(Listos et al. 2019; Creed et al. 2014; Xi 2002). By attaching to the μ-receptors, the
exogenous opioids like morphine and heroine decrease the amount of GABA
(a neurotransmitter) released. More often than not, GABA reduces the amount of
dopamine released in the nucleus accumbens (NAcc). Hence, by inhibiting GABA,
the opiates eventually increase the concentration of dopamine produced and conse-
quently the amount of pleasure felt (Dubhashi 2018; Nuechterlein 2016; Shirayama
and Chaki 2006). Opiates also have dopamine-independent effects within the NAcc,
which play an important role in opiate reward (Ting-A-Kee and Van Der Kooy 2012;
Tomkins and Sellers 2001; O’malley et al. 1992; Shippenberg and Elmer 1998;
Koob and Bloom 1988).
Besides this, the periaqueductal gray (PAG) in the midbrain region being rich in
endogenous opioids and opioid receptors is a major target of analgesic action in CNS
(Tsagareli et al. 2012; Pathan and Williams 2012; Mansour et al. 1995). The
analgesic action of opioids on PAG is exerted by the suppression of inhibitory
402 M. N. Al–Qattan et al.
12.6 Conclusion
Opioid peptides are endogenous ligands for opioid receptors. Proteolytic processing
of larger precursor proteins generates the peptides. The peptides are stored in dense
vesicles within neurons and released upon activation. The release is not restricted to
synaptic space; thus peptides may signal other neurons by volume transmission.
Opioid peptides inhibit the release of neurotransmitters by the affected neurons, thus
modulating their signal propagations. The action of opioid peptides is mediated by
binding GPCR group of receptors (opioid receptors) by binding to the orthosteric
binding site. However, the affinity/efficacy of orthosteric ligand is affected by
cooperation with ligand at allosteric site. Interestingly, many orthosteric and alloste-
ric ligands show biased activation of intracellular second messengers, which
12 Pharmacology of Endogenous Opioids, Opiates and Their Receptors 403
12.7 Remark
The authors declare that theory of biological evolution and its related terms men-
tioned in this chapter and in references are not considered per se by them.
References
Agnati LF, Bjelke B, Fuxe K (1995) Volume versus wiring transmission in the brain: a new
theoretical frame for neuropsychopharmacology. Med Res Rev 15:33–45
Al-Hasani R, Bruchas MR (2011) Molecular mechanisms of opioid receptor-dependent signaling
and behavior. Anesthesiology 115:1363–1381
Allouche SP, Noble F, Marie N (2014) Opioid receptor desensitization: mechanisms and its link to
tolerance. Front Pharmacol 5:280
Asvadi NH, Morgan M, Herath HM, Hewavitharana AK, Shaw PN, Cabot PJ (2014a) Beta-
endorphin 1-31 biotransformation and cAMP modulation in inflammation. PLoS One 9:e90380
Asvadi NH, Morgan M, Hewavitharana AK, Shaw PN, Cabot PJ (2014b) Biotransformation of
beta-endorphin and possible therapeutic implications. Front Pharmacol 5:18–18
Bailey CR, Cordell E, Sobin SM, Neumeister A (2013) Recent progress in understanding the
pathophysiology of post-traumatic stress disorder. CNS Drugs 27:221–232
Basbaum AI, Fields HL (1984) Endogenous pain control systems: brainstem spinal pathways and
endorphin circuitry. Annu Rev Neurosci 7:309–338
Bennett GJ (2000) Update on the neurophysiology of pain transmission and modulation: focus on
the NMDA-receptor. J Pain Symptom Manage 19:2–6
Bermudez M, Nguyen TN, Omieczynski C, Wolber G (2019) Strategies for the discovery of biased
GPCR ligands. Drug Discov Today 24:1031–1037
Berrocoso E, Mico J-A (2009) Cooperative opioid and serotonergic mechanisms generate superior
antidepressant-like effects in a mice model of depression. Int J Neuropsychopharmacol
12:1033–1044
Bie B (2005) cAMP-mediated mechanisms for pain sensitization during opioid withdrawal. J
Neurosci 25:3824–3832
Bisson JI, Cosgrove S, Lewis C, Roberts NP (2015) Post-traumatic stress disorder. BMJ 351:h6161
Bodnar RJ (2018) Endogenous opiates and behavior: 2016. Peptides 101:167–212
Bohn LM, Lefkowitz RJ, Gainetdinov RR, Peppel K, Caron MG, Lin FT (1999) Enhanced
morphine analgesia in mice lacking beta-arrestin 2. Science 286:2495–2498
Bohn LM, Gainetdinov RR, Lin FT, Lefkowitz RJ, Caron MG (2000) Mu-opioid receptor desensi-
tization by beta-arrestin-2 determines morphine tolerance but not dependence. Nature
408:720–723
Bologna Z, Teoh JP, Bayoumi AS, Tang Y, Kim IM (2017) Biased G protein-coupled receptor
signaling: new player in modulating physiology and pathology. Biomol Ther (Seoul) 25:12–25
Browne CA, Lucki I (2019) Targeting opioid dysregulation in depression for the development of
novel therapeutics. Pharmacol Ther. https://doi.org/10.1016/j.pharmthera.2019.04.009
Brownstein MJ (1993) A brief history of opiates, opioid peptides, and opioid receptors. Proc Natl
Acad Sci U S A 90:5391–5393
404 M. N. Al–Qattan et al.
Brust TF, Morgenweck J, Kim SA, Rose JH, Locke JL, Schmid CL, Zhou L, Stahl EL, Cameron
MD, Scarry SM, AubÉ J, Jones SR, Martin TJ, Bohn LM (2016) Biased agonists of the kappa
opioid receptor suppress pain and itch without causing sedation or dysphoria. Sci Signal 9:ra117
Burford NT, Livingston KE, Canals M, Ryan MR, Budenholzer LM, Han Y, Shang Y, Herbst JJ,
O’connell J, Banks M, Zhang L, Filizola M, Bassoni DL, Wehrman TS, Christopoulos A,
Traynor JR, Gerritz SW, Alt A (2015) Discovery, synthesis, and molecular pharmacology of
selective positive allosteric modulators of the delta-opioid receptor. J Med Chem 58:4220–4229
Butt AM, Kalsi A (2006) Inwardly rectifying potassium channels (Kir) in central nervous system
glia: a special role for Kir4.1 in glial functions. J Cell Mol Med 10:33–44
Cai Z, Ratka A (2012) Opioid system and Alzheimer’s disease. Neuromol Med 14:91–111
Calo G, Guerrini R, Rizzi A, Salvadori S, Regoli D (2000) Pharmacology of nociceptin and its
receptor: a novel therapeutic target. Br J Pharmacol 129:1261–1283
Carlezon WA, Krystal AD (2016) Kappa-opioid antagonists for psychiatric disorders: from bench
to clinical trials. Depress Anxiety 33:895–906
Castro MG, Morrison E (1997) Post-translational processing of proopiomelanocortin in the pitui-
tary and in the brain. Crit Rev Neurobiol 11:35–37
Catterall WA (2011) Voltage-gated calcium channels. Cold Spring Harb Perspect Biol 3:a003947–
a003947
Catterall WA, Leal K, Nanou E (2013) Calcium channels and short-term synaptic plasticity. J Biol
Chem 288:10742–10749
Cawley NX, Li Z, Loh YP (2016) 60 years of POMC: biosynthesis, trafficking, and secretion of
pro-opiomelanocortin-derived peptides. J Mol Endocrinol 56:T77–T97
Chattopadhyay M, Mata M, Fink DJ (2008) Continuous δ-opioid receptor activation reduces
neuronal voltage-gated sodium channel (NaV1. 7) levels through activation of protein kinase
C in painful diabetic neuropathy. J Neurosci 28:6652–6658
Chaudhary AMD, Khan MF, Dhillon SS, Naveed S (2019) A review of samidorphan: a novel
opioid antagonist. Cureus. https://doi.org/10.7759/cureus.5139
Chavkin C, Koob GF (2016) Dynorphin, dysphoria, and dependence: the stress of addiction.
Neuropsychopharmacology 41:373–374
Che T, Majumdar S, Zaidi SA, Ondachi P, Mccorvy JD, Wang S, Mosier PD, Uprety R, Vardy E,
Krumm BE, Han GW, Lee MY, Pardon E, Steyaert J, Huang XP, Strachan RT, Tribo AR,
Pasternak GW, Carroll FI, Stevens RC, Cherezov V, Katritch V, Wacker D, Roth BL (2018)
Structure of the nanobody-stabilized active state of the kappa opioid receptor. Cell 172:55–67.
e15
Chieng B, Bekkers JM (2001) Inhibition of calcium channels by opioid-and adenosine-receptor
agonists in neurons of the nucleus accumbens. Br J Pharmacol 133:337–344
Clark SD, Abi-Dargham A (2019) The role of dynorphin and the kappa opioid receptor in the
symptomatology of schizophrenia: a review of the evidence. Biol Psychiatry 86:502–511
Coimbra JRM, Marques DFF, Baptista SJ, Pereira CMF, Moreira PI, Dinis TCP, Santos AE,
Salvador JAR (2018) Highlights in BACE1 inhibitors for Alzheimer’s disease treatment.
Front Chem 6:178
Collard MW, Day R, Akil H, Uhler MD, Douglass JO (1990) Sertoli cells are the primary site of
prodynorphin gene expression in rat testis: regulation of mRNA and secreted peptide levels by
cyclic adenosine 3’,5’-monophosphate analogs in cultured cells. Mol Endocrinol 4:1488–1496
Commons KG (2010) Neuronal pathways linking substance P to drug addiction and stress. Brain
Res 1314:175–182
Conibear AE, Kelly E (2019) A biased view of mu-opioid receptors? Mol Pharmacol 96:542–549
Connor M, Christie MJ (1999) Opioid receptor signalling mechanisms. Clin Exp Pharmacol Physiol
26:493–499
Corder G, Castro DC, Bruchas MR, Scherrer G (2018) Endogenous and exogenous opioids in pain.
Annu Rev Neurosci. https://doi.org/10.1146/annurev-neuro-080317-061522
12 Pharmacology of Endogenous Opioids, Opiates and Their Receptors 405
Egleton RD, Mitchell SA, Huber JD, Janders J, Stropova D, Polt R, Yamamura HI, Hruby VJ,
Davis TP (2000) Improved bioavailability to the brain of glycosylated Met-enkephalin analogs.
Brain Res 881:37–46
Ehrich E, Turncliff R, Du Y, Leigh-Pemberton R, Fernandez E, Jones R, Fava M (2014) Evaluation
of opioid modulation in major depressive disorder. Neuropsychopharmacology 40:1448–1455
Elman I, Borsook D (2019) The failing cascade: comorbid post traumatic stress- and opioid use
disorders. Neurosci Biobehav Rev 103:374–383
Evans RM, You H, Hameed S, Altier C, Mezghrani A, Bourinet E, Zamponi GW (2010)
Heterodimerization of ORL1 and opioid receptors and its consequences for N-type calcium
channel regulation. J Biol Chem 285:1032–1040
Feng Y, He X, Yang Y, Chao D, Lazarus LH, Xia Y (2012) Current research on opioid receptor
function. Current Drug Targets 13:230–246
Fields HL, Margolis EB (2015) Understanding opioid reward. Trends Neurosci 38:217–225
Finnegan TF, Chen S-R, Pan H-L (2006) μ opioid receptor activation inhibits GABAergic inputs to
basolateral amygdala neurons through Kv1.1/1.2 channels. J Neurophysiol 95:2032–2041
Froehlich JC (1997) Opioid peptides. Alcohol health Res World 21:132–135
Gendron L, Mittal N, Beaudry H, Walwyn W (2015) Recent advances on the δ opioid receptor:
from trafficking to function. Br J Pharmacol 172:403–419
Geracioti TD, Strawn JR, Ekhator NN, Wortman M, Kasckow J (2009) 82 - Neuroregulatory
peptides of central nervous system origin: from laboratory to clinic. In: PFAFF DW, Arnold AP,
Etgen AM, Fahrbach SE, Rubin RT (eds) Hormones, brain and behavior, 2nd edn. Academic
Press, San Diego
Ghelardini C, Mannelli LDC, Bianchi E (2015) The pharmacological basis of opioids. Clin Cases
Miner Bone Metab 12:219
Gill BJA, Khan FA, Mckhann GM (2018) You’re not hallucinating: potential new targets for
schizophrenia treatment. Neurosurgery 84:E146–E147
Granier S, Manglik A, Kruse AC, Kobilka TS, Thian FS, Weis WI, Kobilka BK (2012) Structure of
the delta-opioid receptor bound to naltrindole. Nature 485:400–404
Greenwald JD, Shafritz KM (2018) An integrative neuroscience framework for the treatment of
chronic pain: from cellular alterations to behavior. Front Integr Neurosci 12:18
Groer CE, Tidgewell K, Moyer RA, Harding WW, Rothman RB, Prisinzano TE, Bohn LM (2007)
An opioid agonist that does not induce mu-opioid receptor – arrestin interactions or receptor
internalization. Mol Pharmacol 71:549–557
Gu ZH, Wang B, Kou ZZ, Bai Y, Chen T, Dong YL, Li H, Li YQ (2017) Endomorphins: promising
endogenous opioid peptides for the development of novel analgesics. Neurosignals 25:98–116
Guerrero M, Urbano M, Kim E-K, Gamo AM, Riley S, Abgaryan L, Leaf N, Van Orden LJ, Brown
SJ, Xie JY, Porreca F, Cameron MD, Rosen H, Roberts E (2019) Design and synthesis of a
novel and selective kappa opioid receptor (KOR) antagonist (Btrx-335140). J Med Chem
62:1761–1780
Günther T, Dasgupta P, Mann A, Miess E, Kliewer A, Fritzwanker S, Steinborn R, Schulz S (2017)
Targeting multiple opioid receptors – improved analgesics with reduced side effects? Br J
Pharmacol 175:2857–2868
Hassan AN, Foll BL, Imtiaz S, Rehm J (2017) The effect of post-traumatic stress disorder on the
risk of developing prescription opioid use disorder: results from the National Epidemiologic
Survey on Alcohol and Related Conditions III. Drug Alcohol Depend 179:260–266
Heinke B, Gingl E, SandkÜhler J (2011) Multiple targets of μ-opioid receptor-mediated presynaptic
inhibition at primary afferent Aδ- and C-fibers. J Neurosci 31:1313–1322
Herath HMDR, Cabot PJ, Shaw PN, Hewavitharana AK (2012) Study of beta endorphin metabo-
lism in inflamed tissue, serum and trypsin solution by liquid chromatography–tandem mass
spectrometric analysis. Anal Bioanal Chem 402:2089–2100
Hersh LB, Rodgers DW (2008) Neprilysin and amyloid beta peptide degradation. Curr Alzheimer
Res 5:225–231
12 Pharmacology of Endogenous Opioids, Opiates and Their Receptors 407
Ho JH, Stahl EL, Schmid CL, Scarry SM, Aube J, Bohn LM (2018) G protein signaling-biased
agonism at the kappa-opioid receptor is maintained in striatal neurons. Sci Signal 11. https://doi.
org/10.1126/scisignal.aar4309
Hökfelt T, Broberger C, Xu Z-QD, Sergeyev V, Ubink R, Diez M (2000) Neuropeptides — an
overview. Neuropharmacology 39:1337–1356
Holzer P (2009) Opioid receptors in the gastrointestinal tract. Regul Pept 155:11–17
Huang W, Manglik A, Venkatakrishnan AJ, Laeremans T, Feinberg EN, Sanborn AL, Kato HE,
Livingston KE, Thorsen TS, Kling RC, Granier S, Gmeiner P, Husbands SM, Traynor JR, Weis
WI, Steyaert J, Dror RO, Kobilka BK (2015) Structural insights into micro-opioid receptor
activation. Nature 524:315–321
Hughes J, Smith T, Morgan B, Fothergill L (1975) Purification and properties of enkephalin — The
possible endogenous ligand for the morphine receptor. Life Sci 16:1753–1758
Hughes J, Beaumont A, Fuentes JA, Malfroy B, Unsworth C (1980) Opioid peptides: aspects of
their origin, release and metabolism. J Exp Biol 89:239–255
Hurd YL (2002) Subjects with major depression or bipolar disorder show reduction of
prodynorphin mRNA expression in discrete nuclei of the amygdaloid complex. Mol Psychiatry
7:75–81
Ingram SL, Vaughan CW, Bagley EE, Connor M, Christie MJ (1998) Enhanced Opioid Efficacy in
Opioid Dependence Is Caused by an Altered Signal Transduction Pathway. J Neurosci
18:10269–10276
Iwaszkiewicz KS, Schneider JJ, Hua S (2013) Targeting peripheral opioid receptors to promote
analgesic and anti-inflammatory actions. Front Pharmacol 4:132
Jeon HJ, Baek JH, Ahn Y-M, Kim SJ, Ha TH, Cha B, Moon E, Kang H-J, Ryu V, Cho C-H, Heo
J-Y, Kim K, Lee H-J (2016) Review of cohort studies for mood disorders. Psychiatry Investig
13:265
Ji H, Wang Y, Liu G, Chang L, Chen Z, Zhou D, Xu X, Cui W, Hong Q, Jiang L, Li J, Zhou X,
Li Y, Guo Z, Zha Q, Niu Y, Weng Q, Duan S, Wang Q (2017) Elevated OPRD1 promoter
methylation in Alzheimer’s disease patients. PLoS One 12:e0172335
Johnston TH, Versi E, Howson PA, Ravenscroft P, Fox SH, Hill MP, Reidenberg BE, Corey R,
Brotchie JM (2018) DPI-289, a novel mixed delta opioid agonist / mu opioid antagonist
(DAMA), has L-DOPA-sparing potential in Parkinson’s disease. Neuropharmacology
131:116–127
Kalia LV, Lang AE (2015) Parkinson’s disease. Lancet 386:896–912
Kerage D, Sloan EK, Mattarollo SR, Mccombe PA (2019) Interaction of neurotransmitters and
neurochemicals with lymphocytes. J Neuroimmunol 332:99–111
Khan SM, Sleno R, Gora S, Zylbergold P, Laverdure J-P, LabbÉ J-C, Miller GJ, HÉbert TE (2013)
The expanding roles of Gβγ subunits in G protein–coupled receptor signaling and drug action.
Pharmacol Rev 65:545–577
Knoll AT, Carlezon WA (2010) Dynorphin, stress, and depression. Brain Res 1314:56–73
Koehl A, Hu H, Maeda S, Zhang Y, Qu Q, Paggi JM, Latorraca NR, Hilger D, Dawson R, Matile H,
Schertler GFX, Granier S, Weis WI, Dror RO, Manglik A, Skiniotis G, Kobilka BK (2018)
Structure of the micro-opioid receptor-Gi protein complex. Nature 558:547–552
Koob GF, Bloom FE (1988) Cellular and molecular mechanisms of drug dependence. Science
242:715–723
Kosten T, George T (2002) The neurobiology of opioid dependence: implications for treatment. Sci
Pract Perspect 1:13–20
Kosterlitz HW, Hughes J (1975) Some thoughts on the significance of enkephalin, the endogenous
ligand. Life Sci 17:91–96
Kruegel AC, Gassaway MM, Kapoor A, VÁradi A, Majumdar S, Filizola M, Javitch JA, Sames D
(2016) Synthetic and receptor signaling explorations of the mitragyna alkaloids: mitragynine as
an atypical molecular framework for opioid receptor modulators. J Am Chem Soc
138:6754–6764
408 M. N. Al–Qattan et al.
Lacy BE, Mearin F, Chang L, Chey WD, Lembo AJ, Simren M, Spiller R (2016) Bowel disorders.
Gastroenterology 150:1393–1407.e5
Lalanne L, Ayranci G, Kieffer BL, Lutz P-E (2014) The kappa opioid receptor: from addiction to
depression, and back. Front Psychiatry 5:170
Lamberts JT, Jutkiewicz EM, Mortensen RM, Traynor JR (2011) mu-Opioid receptor coupling to
Galpha(o) plays an important role in opioid antinociception. Neuropsychopharmacology
36:2041–2053
Latremoliere A, Woolf CJ (2009) Central sensitization: a generator of pain hypersensitivity by
central neural plasticity. J Pain 10:895–926
Law PY (2011) Opioid receptor signal transduction mechanisms. In: Pasternak GW (ed) The opiate
receptors. Humana Press, Totowa, NJ
Leith JL, Wilson AW, Donaldson LF, Lumb BM (2007) Cyclooxygenase-1-derived prostaglandins
in the periaqueductal gray differentially control C- versus A-fiber-evoked spinal nociception. J
Neurosci 27:11296–11305
Lembeck F (2008) The archeology of substance P. Neuropeptides 42:444–453
Li CH, Chung D (1976) Isolation and structure of an untriakontapeptide with opiate activity from
camel pituitary glands. Proc Natl Acad Sci U S A 73:1145–1148
Li Y, Lefever MR, Muthu D, Bidlack JM, Bilsky EJ, Polt R (2012) Opioid glycopeptide analgesics
derived from endogenous enkephalins and endorphins. Future Med Chem 4:205–226
Li J, Blankenship ML, Baccei ML (2013) Inward-rectifying potassium (Kir) channels regulate
pacemaker activity in spinal nociceptive circuits during early life. J Neurosci 33:3352–3362
Listos J, Łupina M, Talarek S, Mazur A, Orzelska-GÓrka J, KotliŃska J (2019) The mechanisms
involved in morphine addiction: an overview. Int J Mol Sci 20:4302
Livingston KE, Traynor JR (2018) Allostery at opioid receptors: modulation with small molecule
ligands. Br J Pharmacol 175:2846–2856
Livingston KE, Stanczyk MA, Burford NT, Alt A, Canals M, Traynor JR (2018) Pharmacologic
evidence for a putative conserved allosteric site on opioid receptors. Mol Pharmacol 93:157–167
Loh YP, Gainer H (1978) The role of glycosylation on the biosynthesis, degradation, and secretion
of the ACTH-beta-lipotropin common precursor and its peptide products. FEBS Lett
96:269–272
Loose M, Ronnekleiv O, Kelly M (1990) Membrane properties and response to opioids of identified
dopamine neurons in the guinea pig hypothalamus. J Neurosci 10:3627–3634
Luca G, Federico S, Roberto A (2007) Re-discussion of the importance of ionic interactions in
stabilizing ligand-opioid receptor complex and in activating signal transduction. Curr Drug
Targets 8:185–196
Ludwig M, Leng G (2006) Dendritic peptide release and peptide-dependent behaviours. Nat Rev
Neurosci 7:126–136
Lutz PE, Kieffer BL (2013) Opioid receptors: distinct roles in mood disorders. Trends Neurosci
36:195–206
Mabrouk OS, Marti M, Salvadori S, Morari M (2009) The novel delta opioid receptor agonist
UFP-512 dually modulates motor activity in hemiparkinsonian rats via control of the nigro-
thalamic pathway. Neuroscience 164:360–369
Mabrouk OS, Viaro R, Volta M, Ledonne A, Mercuri N, Morari M (2014) stimulation of opioid
receptor and blockade of nociceptin/orphanin FQ receptor synergistically attenuate Parkinson-
ism. J Neurosci 34:12953–12962
Machelska H, Celik MÖ (2018) Advances in achieving opioid analgesia without side effects. Front
Pharmacol 9. https://doi.org/10.3389/fphar.2018.01388
Madariaga-MazÓN A, Marmolejo-Valencia AF, Li Y, Toll L, Houghten RA, Martinez-Mayorga K
(2017) Mu-opioid receptor biased ligands: a safer and painless discovery of analgesics? Drug
Discov Today 22:1719–1729
Mahmod Al-Qattan MN, Mordi MN (2019) Molecular basis of modulating adenosine receptors
activities. Curr Pharm Design 25:817–831
12 Pharmacology of Endogenous Opioids, Opiates and Their Receptors 409
Mains RE, Eipper BA (1981) Coordinate, equimolar secretion of smaller peptide products derived
from pro-ACTH/endorphin by mouse pituitary tumor cells. J Cell Biol 89:21–28
Malfroy B, Swerts JP, Guyon A, Roques BP, Schwartz JC (1978) High-affinity enkephalin-
degrading peptidase in brain is increased after morphine. Nature 276:523–526
Manglik A, Kruse AC, Kobilka TS, Thian FS, Mathiesen JM, Sunahara RK, Pardo L, Weis WI,
Kobilka BK, Granier S (2012) Crystal structure of the micro-opioid receptor bound to a
morphinan antagonist. Nature 485:321–326
Manglik A, Lin H, Aryal DK, Mccorvy JD, Dengler D, Corder G, Levit A, Kling RC, Bernat V,
Hubner H, Huang XP, Sassano MF, Giguere PM, Lober S, Da D, Scherrer G, Kobilka BK,
Gmeiner P, Roth BL, Shoichet BK (2016) Structure-based discovery of opioid analgesics with
reduced side effects. Nature 537:185–190
Mansour A, Fox CA, Akil H, Watson SJ (1995) Opioid-receptor mRNA expression in the rat CNS:
anatomical and functional implications. Trends Neurosci 18:22–29
Markman J, Gudin J, Rauck R, Argoff C, Rowbotham M, Agaiby E, Gimbel J, Katz N, Doberstein
SK, Tagliaferri M, Lu L, Siddhanti S, Hale M (2019) SUMMIT-07. Pain 160:1374–1382
Martin WF, Correll CU, Weiden PJ, Jiang Y, Pathak S, Dipetrillo L, Silverman BL, Ehrich EW
(2019) Mitigation of olanzapine-induced weight gain with samidorphan, an opioid antagonist: a
randomized double-blind phase 2 study in patients with schizophrenia. Am J Psychiatry
176:457–467
Mathieu-Kia A-M, Fan L-Q, Kreek MJ, Simon EJ, Hiller JM (2001) μ-, δ- and κ-opioid receptor
populations are differentially altered in distinct areas of postmortem brains of Alzheimer’s
disease patients. Brain Res 893:121–134
Matsushima A, Sese J, Koyanagi KO (2019) Biosynthetic short neuropeptides: a rational theory
based on experimental results for the missing pain-relief opioid endomorphin precursor gene.
Chembiochem 20:2054–2058
Maves TJ, Pechman PS, Meller ST, Gebhart G (1994) Ketorolac potentiates morphine
antinociception during visceral nociception in the rat. Anesthesiology 80:1094–1101
McCarty D, Priest KC, Korthuis PT (2018) Treatment and prevention of opioid use disorder:
challenges and opportunities. Annu Rev Public Health 39:525–541
McDonald J, Lambert D (2005) Opioid receptors. Contin Educ Anaesth Crit Care Pain 5:22–25
Meguro Y, Miyano K, Hirayama S, Yoshida Y, Ishibashi N, Ogino T, Fujii Y, Manabe S, Eto M,
Nonaka M, Fujii H, Ueta Y, Narita M, Sata N, Yada T, Uezono Y (2018) Neuropeptide oxytocin
enhances μ opioid receptor signaling as a positive allosteric modulator. J Pharmacol Sci
137:67–75
Meilandt WJ, Yu G-Q, Chin J, Roberson ED, Palop JJ, Wu T, Scearce-Levie K, Mucke L (2008)
Enkephalin elevations contribute to neuronal and behavioral impairments in a transgenic mouse
model of Alzheimer’s disease. J Neurosci 28:5007–5017
Mello NK, Negus SS (2006) Interactions between kappa opioid agonists and cocaine: preclinical
studies. Annal N Y Acad Sci 909:104–132
Ménard C, Herzog H, Schwarzer C, Quirion R (2013) Possible role of dynorphins in Alzheimer’s
disease and age-related cognitive deficits. Neurodegener Dis 13:82–85
Mendell LM (2011) Computational functions of neurons and circuits signaling injury: relationship
to pain behavior. Proc Natl Acad Sci U S A 108:15596–15601
Metzger TG, Ferguson DM (1995) On the role of extracellular loops of opioid receptors in
conferring ligand selectivity. FEBS Lett 375:1–4
Mika J, Obara I, Przewlocka B (2011) The role of nociceptin and dynorphin in chronic pain:
implications of neuro-glial interaction. Neuropeptides 45:247–261
Mitznegg P, Domschke W, Sprugel W, Domschke S, Subramanian N, Wunsch E, Moroder L,
Demling L (1977) Enkephalins inhibit intestinal motility: mode of action. Acta
Hepatogastroenterol (Stuttg) 24:119–120
Mogil JS, Pasternak GW (2001) The molecular and behavioral pharmacology of the orphanin
Fq/nociceptin peptide and receptor family. Pharmacol Rev 53:381–415
410 M. N. Al–Qattan et al.
Mores KL, Cummins BR, Cassell RJ, Van Rijn RM (2019) A review of the therapeutic potential of
recently developed G protein-biased kappa agonists. Front Pharmacol 10
Motel CW, Coop A, Cunningham CW (2013) Cholinergic modulation by opioid receptor ligands:
potential application to Alzheimer’s disease. Mini Rev Med Chem 13:456–466
Nandhu MS, Naijil G, Smijin S, Jayanarayanan S, Paulose CS (2010) Opioid system functional
regulation in neurological disease management. J Neurosci Res 88:3215–3221
Neves SR, Ram PT, Iyengar R (2002) G protein pathways. Science 296:1636–1639
Niciu MJ, Arias AJ (2013) Targeted opioid receptor antagonists in the treatment of alcohol use
disorders. CNS Drugs 27:777–787
Nieto MM, Guen SL, Kieffer BL, Roques BP, Noble F (2005) Physiological control of emotion-
related behaviors by endogenous enkephalins involves essentially the delta opioid receptors.
Neuroscience 135:305–313
Norn S, Kruse PR, Kruse E (2005) History of opium poppy and morphine. Dan Medicinhist Arbog
33:171–184
North R (1993) Opioid actions on membrane ion channels. In: Opioids. Springer, New York, NY
North RA, Williams JT (1983) How do opiates inhibit neurotransmitter release? Trends Neurosci
6:337–339
Nuechterlein EB (2016) Alterations in endogenous opioid neurotransmission associated with acute
and long-term use of drugs of abuse. PhD Thesis, University of Michigan, Ann Arbor, USA.
https://deepblue.lib.umich.edu/bitstream/handle/2027.42/120787/emilybn_1.pdf?sequence¼1
O’malley SS, Jaffe AJ, Chang G, Schottenfeld RS, Meyer RE, Rounsaville B (1992) Naltrexone
and coping skills therapy for alcohol dependence: a controlled study. Arch Gen Psychiatry
49:881–887
Ohbuchi K, Miyagi C, Suzuki Y, Mizuhara Y, Mizuno K, Omiya Y, Yamamoto M, Warabi E,
Sudo Y, Yokoyama A, Miyano K, Hirokawa T, Uezono Y (2016) Ignavine: a novel allosteric
modulator of the μ opioid receptor. Sci Rep 6:31748
Owen MJ, Sawa A, Mortensen PB (2016) Schizophrenia. Lancet 388:86–97
Pan J, Cai H (2017) Opioid system in L-Dopa-induced dyskinesia. Transl Neurodegener. https://
deepblue.lib.umich.edu/bitstream/handle/2027.42/120787/emilybn_1.pdf?sequence¼1
Papadimitriou A, Priftis KN (2009) Regulation of the hypothalamic-pituitary-adrenal axis.
Neuroimmunomodulation 16:265–271
Patel RM, Shulman ST (2015) Kawasaki disease: a comprehensive review of treatment options. J
Clin Pharm Ther 40:620–625
Pathan H, Williams J (2012) Basic opioid pharmacology: an update. Br J Pain 6:11–16
Pert CB, Snyder SH (1973) Properties of opiate-receptor binding in rat brain. Proc Natl Acad Sci U
S A 70:2243–2247
Petrenko AB, Yamakura T, Baba H, Shimoji K (2003) The role of N-methyl-D-aspartate (NMDA)
receptors in pain: a review. Anesth Analg 97:1108–1116
Pleuvry BJ (1991) Opioid receptors and their ligands: natural and unnatural. Br J Anaesth
66:370–380
Podvin S, Yaksh T, Hook V (2016) The emerging role of spinal dynorphin in chronic pain: a
therapeutic perspective. Annu Rev Pharmacol Toxicol 56:511–533
Polo S, DÍaz AF, Gallardo N, LeÁnez S, Balboni G, Pol O (2019) Treatment with the delta opioid
agonist Ufp-512 alleviates chronic inflammatory and neuropathic pain: mechanisms implicated.
Front Pharmacol 10:283
Portoghese PS, Sultana M, Nagase H, Takemori AE (1988) Application of the message-address
concept in the design of highly potent and selective non-peptide .delta. opioid receptor
antagonists. J Med Chem 31:281–282
Przewlocki R (2013) Opioid peptides. In: Pfaff DW (ed) Neuroscience in the 21st century: from
basic to clinical. New York, NY, Springer
Ramaswamy S, Langford R (2017) Antinociceptive and immunosuppressive effect of opioids in an
acute postoperative setting: an evidence-based review. BJA Educ 17:105–110
12 Pharmacology of Endogenous Opioids, Opiates and Their Receptors 411
Ranjan R, Pandey S, Shukla AK (2017) Biased opioid receptor ligands: gain without pain. Trends
Endocrinol Metab 28:247–249
Reindl JD, Rowan K, Carey AN, Peng X, Neumeyer JL, McLaughlin JP (2008) Antidepressant-like
effects of the novel kappa opioid antagonist MCL-144B in the forced-swim test. Pharmacology
81:229–235
Reisine T, Bell GI (1993) Molecular biology of opioid receptors. Trends Neurosci 16:506–510
Rodrı́guez-Manzo G, Asai M, Fernández-Guasti A (2002) Evidence for changes in brain enkephalin
contents associated to male rat sexual activity. Behav Brain Res 131:47–55
Roth BL, Baner K, Westkaemper R, Siebert D, Rice KC, Steinberg S, Ernsberger P, Rothman RB
(2002) Salvinorin A: a potent naturally occurring nonnitrogenous opioid selective agonist. Proc
Natl Acad Sci 99:11934–11939
Rubinstein M, Mogil JS, JapÓN M, Chan EC, Allen RG, Low MJ (1996) Absence of opioid stress-
induced analgesia in mice lacking beta-endorphin by site-directed mutagenesis. Proc Natl Acad
Sci 93:3995–4000
Rush AM, Cummins TR, Waxman SG (2007) Multiple sodium channels and their roles in
electrogenesis within dorsal root ganglion neurons. J Physiol 579:1–14
Russo AF (2017) Overview of neuropeptides: awakening the senses? Headache 57(Suppl 2):37–46
Sarajarvi T, Marttinen M, Natunen T, Kauppinen T, MÄkinen P, Helisalmi S, Laitinen M,
Rauramaa T, Leinonen V, PetÄJÄ-Repo U, Soininen H, Haapasalo A, Hiltunen M (2015)
Genetic variation in δ-opioid receptor associates with increased β- and γ-secretase activity in the
late stages of Alzheimer’s disease. J Alzheimer’s Dis 48:507–516
Schaffer CB, Nordahl TE, Schaffer LC, Howe J (2007) Mood-elevating effects of opioid analgesics
in patients with bipolar disorder. J Neuropsychiatry 19:449–452
Schapira AHV, Chaudhuri KR, Jenner P (2017) Non-motor features of Parkinson disease. Nat Rev
Neurosci 18:435–450
Scheuer T (2011) Regulation of sodium channel activity by phosphorylation. Semin Cell Dev Biol
22:160–165
Schuckit MA (2016) Treatment of opioid-use disorders. N Engl J Med 375:357–368
Seseña E, Vega R, Soto E (2014) Activation of μ-opioid receptors inhibits calcium-currents in the
vestibular afferent neurons of the rat through a cAMP dependent mechanism. Front Cell
Neurosci 8:90
Sgroi S, Tonini R (2018) Opioidergic modulation of striatal circuits, implications in Parkinson’s
disease and levodopa induced dyskinesia. Front Neurol 9:524
Shalev A, Liberzon I, Marmar C (2017) Post-traumatic stress disorder. N Engl J Med
376:2459–2469
Sheffler DJ, Roth BL (2003) Salvinorin A: the ‘magic mint’ hallucinogen finds a molecular target in
the kappa opioid receptor. Trends Pharmacol Sci 24:107–109
Shekhar A (2019) Role of kappa opioid receptors in symptoms of schizophrenia: what is the
neurobiology? Biol Psychiatry 86:494–496
Sher L (2004) The role of endogenous opioids in the placebo effect in post-traumatic stress disorder.
Complement Med Res 11:354–359
Shippenberg TS, Elmer GI (1998) The neurobiology of opiate reinforcement. Crit Rev Neurobiol
12:267–303
Shirayama Y, Chaki S (2006) Neurochemistry of the nucleus accumbens and its relevance to
depression and antidepressant action in rodents. Curr Neuropharmacol 4:277–291
Smrcka A (2008) G protein βγ subunits: central mediators of G protein-coupled receptor signaling.
Cell Mol Life Sci 65:2191–2214
Snyder SH (2004) Opiate receptors and beyond: 30 years of neural signaling research. Neurophar-
macology 47:274–285
Sprouse-Blum AS, Smith G, Sugai D, Parsa FD (2010) Understanding endorphins and their
importance in pain management. Hawaii Med J 69:70–71
412 M. N. Al–Qattan et al.
Stanczyk MA, Livingston KE, Chang L, Weinberg ZY, Puthenveedu MA, Traynor JR (2019) The
delta-opioid receptor positive allosteric modulator BMS 986187 is a G-protein-biased allosteric
agonist. Br J Pharmacol 176:1649–1663
Takahashi A (2016) Subchapter 7A - enkephalin. In: TAKEI Y, Ando H, Tsutsui K (eds) Handbook
of hormones. Academic Press, San Diego
Takeuchi M (2001) The mammalian pars intermedia—structure and function. Zoolog Sci 18
(133-144):12
Tejeda HA, Shippenberg TS, Henriksson R (2011) The dynorphin/κ-opioid receptor system and its
role in psychiatric disorders. Cell Mol Life Sci 69:857–896
Terskiy A, Wannemacher KM, Yadav PN, Tsai M, Tian B, Howells RD (2007) Search of the
human proteome for endomorphin-1 and endomorphin-2 precursor proteins. Life Sci
81:1593–1601
Theriot J, Azadfard M, Kum B (2019) Opioid antagonists. In: StatPearls. StatPearls Publishing,
Treasure Island (FL)
Thompson GL, Lane JR, Coudrat T, Sexton PM, Christopoulos A, Canals M (2015) Biased
agonism of endogenous opioid peptides at the mu-opioid receptor. Mol Pharmacol 88:335–346
Ting-A-Kee R, Van Der Kooy D (2012) The neurobiology of opiate motivation. Cold Spring Harb
Perspect Med 2:a012096–a012096
Tomkins DM, Sellers EM (2001) Addiction and the brain: the role of neurotransmitters in the cause
and treatment of drug dependence. CMAJ 164:817–821
Torres-Berrio A, Nava-Mesa MO (2019) The opioid system in stress-induced memory disorders:
from basic mechanisms to clinical implications in post-traumatic stress disorder and
Alzheimer’s disease. Prog Neuropsychopharmacol Biol Psychiatry 88:327–338
Tortorici V, Vanegas H (1995) Anti-nociception induced by systemic or PAG-microinjected lysine-
acetylsalicylate in rats. Effects on tail-flick related activity of medullary off-and on-cells. Eur J
Neurosci 7:1857–1865
Tsagareli MG, Tsiklauri N, Nozadze I, Gurtskaia G (2012) Tolerance effects of non-steroidal anti-
inflammatory drugs microinjected into central amygdala, periaqueductal grey, and nucleus
raphe. Neural Regen Res 7:1029–1039
Turner AJ (2004) 108 - Neprilysin. In: Barrett AJ, Rawlings ND, Woessner JF (eds) Handbook of
proteolytic enzymes, 2nd edn. Academic Press, London
van den Pol AN (2012) Neuropeptide transmission in brain circuits. Neuron 76:98–115
Vanderah TW, Ossipov MH, Lai J, Malan PTJ, Porreca F (2001) Mechanisms of opioid-induced
pain and antinociceptive tolerance: descending facilitation and spinal dynorphin. Pain 92:5–9
Varamini P, Mansfeld FM, Blanchfield JT, Wyse BD, Smith MT, Toth I (2012) Synthesis and
biological evaluation of an orally active glycosylated endomorphin-1. J Med Chem
55:5859–5867
Vaughan CW (1998) Enhancement of opioid inhibition of GABAergic synaptic transmission by
cyclo-oxygenase inhibitors in rat periaqueductal grey neurones. Br J Pharmacol 123:1479–1481
Vaughan C, Christie M (1997) Presynaptic inhibitory action of opioids on synaptic transmission in
the rat periaqueductal grey in vitro. J Physiol 498:463–472
Vaughan CW, Ingram SL, Connor MA, Christie MJ (1997) How opioids inhibit GABA-mediated
neurotransmission. Nature 390:611–614
Veening JG, Barendregt HP (2015) The effects of beta-endorphin: state change modification. Fluids
Barriers CNS 12:3–3
Veening JG, Gerrits PO, Barendregt HP (2012) Volume transmission of beta-endorphin via the
cerebrospinal fluid; a review. Fluids Barriers CNS 9:16–16
Venkatraman A, Edlow BL, Immordino-Yang MH (2017) The brainstem in emotion: a review.
Front Neuroanat 11:15
Vergura R, Balboni G, Spagnolo B, Gavioli E, Lambert DG, McDonald J, Trapella C, Lazarus LH,
Regoli D, Guerrini R, Salvadori S, CalÓ G (2008) Anxiolytic- and antidepressant-like activities
of H-Dmt-Tic-Nh-CH(CH2-COOH)-Bid (UFP-512), a novel selective delta opioid receptor
agonist. Peptides 29:93–103
12 Pharmacology of Endogenous Opioids, Opiates and Their Receptors 413
Waldhoer M, Bartlett SE, Whistler JL (2004) Opioid receptors. Annu Rev Biochem 73:953–990
Wang H, Subedi K (2020) δ-Opioid receptor as a potential therapeutic target for ischemic stroke.
Neural Regen Res 15:20
Wang H, Wessendorf MW (2001) Equal proportions of small and large DRG neurons express
opioid receptor mRNAs. J Comp Neurol 429:590–600
Wang W, Gu J, Li Y-Q, Tao Y-X (2011) Are voltage-gated sodium channels on the dorsal root
ganglion involved in the development of neuropathic pain? Mol Pain 7:16
Wang Y, Wang Y-X, Liu T, Law P-Y, Loh HH, Qiu Y, Chen H-Z (2014) μ-opioid receptor
attenuates AβOligomers-induced neurotoxicity through mTOR signaling. CNS Neurosci Ther
21:8–14
Williams WA, Grant JE, Winstanley CA, Potenza MN (2008) Current concepts in the classification,
treatment, and modeling of pathological gambling and other impulse control disorders. In:
McArthur RA, Borsini F (eds) Animal and translational models for CNS drug discovery,
Academic Press, San Diego
Wilson AM, Soignier RD, Zadina JE, Kastin AJ, Nores WL, Olson RD, Olson GA (2000)
Dissociation of analgesic and rewarding effects of endomorphin-1 in rats. Peptides
21:1871–1874
Wilson-Poe AR, Jeong H-J, Vaughan CW (2017) Chronic morphine reduces the readily releasable
pool of GABA, a presynaptic mechanism of opioid tolerance. J Physiol 595:6541–6555
Winters BL, Gregoriou GC, Kissiwaa SA, Wells OA, Medagoda DI, Hermes SM, Burford NT,
Alt A, Aicher SA, Bagley EE (2017) Endogenous opioids regulate moment-to-moment neuronal
communication and excitability. Nat Commun 8:14611
Wu ZQ, Chen J, Chi ZQ, Liu JG (2007) Involvement of dopamine system in regulation of Na+,K+-
ATPase in the striatum upon activation of opioid receptors by morphine. Mol Pharmacol
71:519–530
Wu H, Wacker D, Mileni M, Katritch V, Han GW, Vardy E, Liu W, Thompson AA, Huang XP,
Carroll FI, Mascarella SW, Westkaemper RB, Mosier PD, Roth BL, Cherezov V, Stevens RC
(2012) Structure of the human kappa-opioid receptor in complex with JDTic. Nature
485:327–332
Xi Z-X (2002) Gabaergic mechanisms of opiate reinforcement. Alcohol Alcohol 37:485–494
Xia R, Mao Z-H (2012) Progression of motor symptoms in Parkinson’s disease. Neurosci Bull
28:39–48
Xu C, Liu G, Ji H, Chen W, Dai D, Chen Z, Zhou D, Xu L, Hu H, Cui W, Chang L, Zha Q, Li L,
Duan S, Wang Q (2018) Elevated methylation of OPRM1 and OPRL1 genes in Alzheimer’s
disease. Mol Med Rep. https://doi.org/10.3892/mmr.2018.9424
Yakovleva T, Bazov I, Cebers G, Marinova Z, Hara Y, Ahmed A, Vlaskovska M, Johansson B,
Hochgeschwender U, Singh IN, Bruce-Keller AJ, Hurd YL, Kaneko T, Terenius L, EkstrÖM
TJ, Hauser KF, Pickel VM, Bakalkin G (2006) Prodynorphin storage and processing in axon
terminals and dendrites. FASEB J 20:2124–2126
Yakovleva T, Marinova Z, Kuzmin A, Seidah NG, Haroutunian V, Terenius L, Bakalkin G (2007)
Dysregulation of dynorphins in Alzheimer disease. Neurobiol Aging 28:1700–1708
Yam M, Loh Y, Tan C, Khadijah Adam S, Abdul Manan N, Basir R (2018) General pathways of
pain sensation and the major neurotransmitters involved in pain regulation. Int J Mol Sci
19:2164
Yang A, Tsai S-J (2017) New targets for schizophrenia treatment beyond the dopamine hypothesis.
Int J Mol Sci 18:1689
Yehuda R, Hoge CW, McFarlane AC, Vermetten E, Lanius RA, Nievergelt CM, Hobfoll SE,
Koenen KC, Neylan TC, Hyman SE (2015) Post-traumatic stress disorder. Nat Rev Dis Primers
1:15057. https://doi.org/10.1038/nrdp.2015.57
Young J, Mendoza M (2018) Parkinson’s disease: A treatment guide. J Fam Pract 67:276–286
Zadina JE, Hackler L, Ge LJ, Kastin AJ (1997) A potent and selective endogenous agonist for the
mu-opiate receptor. Nature 386:499–502
414 M. N. Al–Qattan et al.
Zamponi GW, Currie KPM (2013) Regulation of CaV2 calcium channels by G protein coupled
receptors. Biochim Biophys Acta 1828:1629–1643
Zamponi GW, Striessnig J, Koschak A, Dolphin AC (2015) The physiology, pathology, and
pharmacology of voltage-gated calcium channels and their future therapeutic Potential.
Pharmacol Rev 67:821–870
Zebraski SE, Kochenash SM, Raffa RB (2000) Lung opioid receptors: pharmacology and possible
target for nebulized morphine in dyspnea. Life Sci 66:2221–2231
Zhang Z, Pan ZZ (2011) Signaling cascades for δ-opioid receptor-mediated inhibition of GABA
synaptic transmission and behavioral antinociception. Mol Pharmacol 81:375–383
Zhang X, Bao L, Arvidsson U, Elde R, HÖkfelt T (1997) Localization and regulation of the delta-
opioid receptor in dorsal root ganglia and spinal cord of the rat and monkey: evidence for
association with the membrane of large dense-core vesicles. Neuroscience 82:1225–1242
Zhao J, Li X, Li B, Chen L, Pei G (2015) A new delta opioid receptor antagonist as a novel drug
against Alzheimer’s disease. Alzheimer’s Dement 11:P371
Zieglgänsberger W (2018) Substance P and pain chronicity. Cell Tissue Res 375:227–241
Pharmacology of Endocannabinoids
and Their Receptors 13
Gaurav Gupta, Wafa Hourani, Pran Kishore Deb, Satyendra Deka,
Pobitra Borah, Juhi Tiwari, Sacchidanand Pathak, and Puneet Kumar
Abstract
G. Gupta · S. Pathak
School of Pharmacy, Suresh Gyan Vihar University, Jaipur, India
W. Hourani (*)
Faculty of Pharmacy, Philadelphia University, Amman, Jordan
e-mail: [email protected]
P. K. Deb
Department of Pharmaceutical Sciences, Faculty of Pharmacy, Philadelphia University, Amman,
Jordan
S. Deka · P. Borah
Pratiksha Institute of Pharmaceutical Sciences, Guwahati, Assam, India
J. Tiwari
Alwar Pharmacy College, Alwar, India
P. Kumar
Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical
University, Bathinda, Punjab, India
among others. This chapter is comprehensively focused on the signal transduction and
metabolic pathways, physiological roles, pharmacology and therapeutic potential of
the endocannabinoids, with particular emphasis on cannabinoid addiction.
Keywords
Abbreviations
Abh4 α/β-Hydrolase 4
2-AG 2-Arachidonoylglycerol
CB Cannabinoid
CB1R Cannabinoid 1 receptor
CB2R Cannabinoid 2 receptor
CBD Cannabidiol
CBN Cannabinol
DAG Diacylglycerol
eCBs Endocannabinoids
FAAH Fatty acid amide hydrolase
GPCRs G protein-coupled receptors
MAGL Monoacylglycerol lipase
MAPK Mitogen-activated protein kinase
NAPE N-arachidonoyl phosphatidylethanolamine
PA2 Phospholipase A2
pCBs Phytocannabinoids
PLA1 PI-explicit phospholipase A1
PLC Phospholipase C
THC Δ9-Tetrahydrocannabinol
13.1 Introduction
The medicinal use of Cannabis sativa or marijuana has a long and rich history dating
back to the sixth century B.C. and it was introduced into western medicine during the
nineteenth century (Gaoni and Mechoulam 1964). Being the oldest source of textile
fibers, cultivation of cannabis originated in Western Asia and Egypt, and later
expanded to Europe and America. Under federal laws in the United States, the
cultivation, use and possession of cannabis is illegal (1982). The legalization of
cannabis for medical and recreational use is supported by many countries, and it has
been legalized in some states of America.
Cannabis sativa is a dioecious species, that is, there is a distinct male and female
flower on separate plants, and it belongs to the Cannabinaceae family (Small et al.
2002). It is generally reported that the psychomimetic property of cannabis is
13 Pharmacology of Endocannabinoids and Their Receptors 417
associated with the female plant and the sticky resins are secreted from the glandular
hair located on the female flowers and their adjacent leaves (Fig. 13.1). A number of
constituents are found in the plant possessing their crucial role in the treatment of
various diseases, among which Δ13-tetrahydrocannabinol (THC) is the most active
constituent (Baron 2018). Cannabis sativa also consists of a hempseed fixed oil
which is found in the seed part of the plant.
This plant is therapeutically indicated in the management of nausea, pain, glau-
coma, neuralgia, cardiovascular disorder, epilepsy, inflammation, cancer, neuropsy-
chological disorder, neurodegenerative diseases, addiction, arthritis, depression and
headache (Adler and Deleo 2019; Akram et al. 2019; Alipour et al. 2019). Recent
data also suggests the use of phytocannabinoids in the management of multiple
sclerosis and HIV/AIDS symptoms.
The compounds identified or isolated from the plant have been continuously
increasing over time. Approximately 565 compounds have been identified from
C. sativa, among which approximately 120 are called cannabinoids. Cannabinoids
(CBs) are the compounds possessing the typical C21 terpenophenolic ring or its
derivatives/transformation products. CBs can be classified into two broad categories
on the basis of the location where they are found (Baron 2018). The CBs found in the
plant are known as phytocannabinoids (pCBs), whereas the CBs obtained from
animals are known as endocannabinoids (eCBs). Phytocannabinoids are either
found in the cannabis plant or in agricultural hemp (Gertsch et al. 2010). CBs are
lipophilic in nature due to which previously it was speculated that the drug directly
disrupts the cellular membrane without any specified pathway. But after the discov-
ery of some phytocannabinoids, the presence of certain receptors was observed
showing an affinity toward the CBs. CBs show their pharmacological effect by
binding with a specific cannabinoid receptor (CBR) found in the animal body, which
can be classified into two types: cannabinoid 1 receptor (CB1R) and cannabinoid
2 receptor (CB2R). These CBRs are G protein-coupled receptors (GPCRs).
Endocannabinoids are lipid-based endogenous cannabimimetic neurotransmitters
which bind to the CBR and CBR-proteins found in both the central nervous system
(CNS) and the peripheral nervous system (PNS) (Andrade et al. 2019; Borsoi et al.
2019; Breit et al. 2019). The eCBs form the endocannabinoid system, which
418 G. Gupta et al.
is involved in the maintenance of the homeostasis of the human body (Battista et al.
2012). The development of the endocannabinoid system depends upon the intake of
nutritional and dietary co-factors. The formation of eCBs occurs according to the
necessity and requirement of the body, and can bind with the receptors after being
produced. The endocannabinoids found in the human body are N-arachidonoyl-
ethanolamine (AEA; anandamide) and 2-arachidonoylglycerol (2-AG). They serve as
the endogenous agonists of CBRs and regulate the CNS as well as the PNS, thus
controlling various physiological functions of the body including the immune system.
Therefore, any changes in the endocannabinoid system will result in various disorders,
from neurodegenerative disorder to arthritis. Homeostasis is maintained due to the
constant enzymatic degradation of eCBs. Anandamide shows higher affinity but less
efficacy toward the CB1R, whereas 2-AG exhibits less affinity but high efficacy for
both the receptors (Carr et al. 2019; Chye et al. 2019; Cohen et al. 2019). The eCBs are
hydrophobic in nature and exhibit slower diffusion. The degradation of eCBs inside
the cell occurs either by oxidation or by hydrolysis. Anandamide is hydrolyzed by
fatty acid amide hydrolase into free arachidonic acid and ethanolamine, whereas 2-AG
is hydrolysed into arachidonic acid and glycerol in the presence of monoacylglycerol
lipase (MAGL). Cyclooxygenase-2 and several lipoxygenases are responsible for
hydrolysing the eCBs by the process of oxidation. Apart from maintaining homeosta-
sis, the eCBs are also responsible for recovery and repair of cells. This may confer
various properties including anti-oxidant, anti-inflammatory, anti-anxiety, anti-psy-
chotic, anti-epilepsy, anti-cancer, anti-nausea, anti-bacterial, anti-diabetic, anti-arthri-
tis, pain relief, bone stimulant, immune modulator, neuroprotective and cardio
protective activities. Some endogenous fatty acid derivatives are also found in the
body, which are known as eCB-like compounds. These substances are known to
promote the activity of classic eCBs by the entourage effect (Dale et al. 2019; Diao and
Huestis 2019; Dinis-Oliveira 2019). Apart from the above two eCBs, other reported
eCBs are noladin ether (2-arachidonylglyceryl ether), arachidonoyl dopamine and
virodhamine. The pharmacological profile and biochemical properties of these eCBs
are not known. This chapter discusses the signal transduction and metabolic pathways,
physiological roles, pharmacology and therapeutic potential of the endocannabinoids.
Cannabinoid receptors are a class of cell membrane receptors that belong to the
family of rhodopsin-like G protein-coupled receptors (GPCR). There are thus two
cannabinoid receptor classes, CB1 and CB2 receptors, which are both coupled to Gi
or Go protein, via negative coupling to adenylyl cyclase (AC) and positive coupling
to the mitogen-activated protein (MAP) kinase family. CB1 receptors are also
coupled to ion channels through the Gi/o proteins. There is precisely positive
coupling to the A-type inward rectifier potassium channels with negative coupling
to N-type, P/Q-type voltage-gated calcium channels and to D-type potassium
channels (Pertwee et al. 2010). Moreover, it is presumed that CB1 also activates
adenylate cyclase types II, IV and VIII via Gsα (Rhee et al. 1998). Both receptors are
13 Pharmacology of Endocannabinoids and Their Receptors 419
Ca+2 K+ Extracellular
CB2
+
-
PKA AKT/PKB mTOR Raf
MEK 1/2
- + +
-/+
Gene expression EKK 1/2
Fig. 13.2 Cannabinoid receptor signaling. CB Cannabinoid receptor, mTOR Mechanistic target of
rapamycin, Akt Protein kinase B, PI3K Phosphatidylinositol-3-kinase, PKA Protein kinase A, ERK
Extracellular signal-regulated kinase
to produce analgesia (Pertwee et al. 2010; Matsuda et al. 1990; Herkenham et al.
1991).
CB2 receptors are predominantly associated with expression in the peripheral
cells derived from the immune system. Expression of the CB2 receptor gene
transcripts was detected in the thymus, mast cells, spleen, tonsils and blood cells
(Galiègue et al. 1995; Munro et al. 1993), where they tend to modulate inflammatory
and immunosuppressive activity and control cytokines release.
In contrast, the CB1 was also detected in several peripheral tissues including the
reproductive system, cardiovascular system and gastrointestinal tract. Recently, the
CB2 was reported to be located in the CNS, for example, in the microglial cells
(Svíženská et al. 2008). It has been demonstrated that CB2Rs expression level in the
cerebrum is much lower than CB1Rs in healthy subjects (Onaivi et al. 2008, 2006;
Nunez et al. 2004). In the brain, CB2A is the significant transcript isoform, while
both CB2A and CB2B transcripts are available in larger amounts in the peripheral
tissue such as spleen, skeletal, cardiovascular, thymus and renal systems (Jordan and
Xi 2019; Rossi et al. 2018) and in immune cells, especially cells of macrophage,
lineage thymus, tonsils, T-lymphocytes, monocytes, natural killer cells, and poly-
morphonuclear cells and B-lymphocytes (Howlett et al. 2002; Schatz et al. 1997;
Galiegue et al. 1995). This prevalent distribution of cannabinoid receptors explains
their wide therapeutic applications in almost every system in the human body.
It has been reported that the CB1 receptor conserves its identity across different
species such as mammals, amphibians and fish (Yamaguchi et al. 1996; Soderstrom
et al. 2000). In contrast, cannabinoid CB2 receptors are more varied. Mukherjee and
co-workers (2004) and Bingham et al. (2007) reported that rat CB2, mouse CB2 and
human CB2 receptors show different pharmacological profiles, although all of them
are considered CB2 receptors (Mukherjee et al. 2004; Bingham et al. 2007). There is
an 81% amino acid sequence in rodent and human CB2 receptors, contrasted with
93% amino acid identity among rodent and mouse CB2 receptors (Griffin et al.
2000).
Notwithstanding CB1 and CB2 receptors, pharmacological studies recommend
the presence of non-CB1, non-CB2 receptors interceding the impacts of
CB. Although a few proteins have been examined as contenders for a potential
“CB3” receptor, the reality is questionable and not yet established (Chen 2016).
13.3.1 Endocannabinoids
This ligand behaves as a CB1 and CB2 receptor partial agonist and demonstrates
higher intrinsic activity toward CB1 than CB2 (Mackie et al. 1993). Gonsiorek and
co-workers reported that anandamide and not its metabolite arachidonic acid
antagonizes hCB2 activation by 2-AG in CHO-hCB2 and anandamide can function
as an endogenous antagonist at the peripheral cannabinoid receptor. This strengthens
the hypothesis that the in vivo immunosuppressive effects of 2-AG or other
cannabinoids are dependent on the local concentration of anandamide and 2-AG
(Gonsiorek et al. 2000).
2-AG was the second endogenous cannabinoid receptor ligand to be discovered
following anandamide (Mechoulam et al. 1995) and better illustrates selective
binding toward CB1 than CB2 and functions as a full agonist. Anandamide and
2-AG show similar affinity toward hCB2. Some other substances have been found to
function as endocannabinoids. These substances comprise N-
dihomo-γ-linolenoylethanolamine, N-docosatetraenoylethanolamine,
arachidonoylethanolamine (virodhamine), oleamide, N-arachidonoyl dopamine
(NADA), Arachidonoyl-serine (ARA-S) and Noladin etherand N-oleoyl dopamine
(for a review, see Pertwee et al. 2010). Table 9.1 represents various eCBs and their
type of interaction with CB1R and CB2R, respectively.
2. 2- 2- Agonist Agonist
Arachidonoylglycerol arachidonoylglycerol
N-Acylphosphatidylethanolamine Diacylglycerol
FAAH MGL
FAAH 2 ABHD6 Hydrolysis
NAAA ABHD12
Fig. 13.3 Schematic representation of the endocannabinoid synthesis and hydrolysis. DAGL
Diacylglycerol lipase, NAPE-PLD N-acylphosphatidylethanolamine phospholipase D, FAAH
fatty acid amide hydrolase, NAAA N-acylethanolamine-hydrolyzing acid amidase, MGL
Monoacylglycerol lipase, ABHD6 α,β-hydrolase 6, ABHD12 α,β-hydrolase 12
by PLC activity and the second step comprises the hydrolysis of DAG by
diacylglycerol (Stella et al. 1997).
opening in the cell membrane, which are associated with the regulation of synapse
discharge (either restraint of glutamate or GABA). Through hindrance of AC and a
decrease of cAMP, or through MAP kinase pathways, eCB could likewise prolong
cell action (Miller and Devi 2011; Tsuboi et al. 2018).
after the past discharge into the extracellular space or straightforwardly moving
inside the cell membrane. Endocannabinoid flagging is restricted by extremely
proficient degradation processes, including encouraging the uptake from the extra-
cellular space into the cell and enzymatic catabolism interceded by explicit intracel-
lular compounds. The molecular nature of the transporter protein(s) associated with
endocannabinoid uptake has not yet been illustrated. Nonetheless, the compounds’
capacity to degrade eCB is quite well characterized (Bara et al. 2018; Baron 2018;
Bonini et al. 2018). They are FAAH for anandamide and related eCBs, and
monoglycerollipase for 2-AG, albeit different chemicals may be in part engaged
with the degradation of this last compound. An intriguing part of endocannabinoid
function is the quick induction of their synthesis, receptor activation and degrada-
tion. The endocannabinoid framework has in this manner been proposed to act on
interest, with a firmly controlled spatial and temporal selectivity. The framework
applies its modulatory activities only when and where it is required. It represents a
significant refinement among physiological elements of the endocannabinoid frame-
work and the biological activities of exogenous CB receptor agonists, which need
such selectivity. With regard to regulation of endocrine, it is intriguing to note here
that hormonal incitement with glucocorticoids can prompt eCB synthesis in the
nerve center by quick nongenomic systems (Bonn-Miller et al. 2018; Bramness et al.
2018; Cardenia et al. 2018). It was likewise demonstrated later that phospholipase C
communicates to an intracellular event finder of membrane depolarization and
receptor incitement, prompting the combination and, potentially, the arrival of
eCB in the hippocampus. This information uncovers a novel component for activa-
tion of the endocannabinoid framework, which could be engaged with the regulation
of endocrine frameworks. This also concerns eCB degradation, which communicates
to a significant regulatory part of the movement of the endocannabinoid framework
(Citti et al. 2018; Colizzi et al. 2018).
13.3.2 Phytocannabinoids
More than 500 compounds have been identified from Cannabis sativa, which are
phytocannabinoids in nature as well as non-phytocannabinoids. A total of
104 phytocannabinoids have been isolated to date, classified into 11 chemical classes
as follows: (–)-delta-9-trans-tetrahydrocannabinol (Δ9-THC), (–)-delta-8-trans-tetra-
hydrocannabinol (Δ8-THC), cannabidiol (CBD), cannabinodiol (CBND),
cannabichromene (CBC), cannabigerol (CBG), cannabinol (CBN), cannabicyclol
(CBL), cannabielsoin (CBE), cannabitriol (CBT) and miscellaneous-type
cannabinoids (Bilbao et al. 2004). Miscellaneous cannabinoids represent compounds
that are composed of different chemical structures and these compounds tend to
differ in their psychoactive properties. In particular, the psychoactive properties of
cannabis have been attributed to the most abundant constituent, Δ9-THC, which was
initially discovered in 1964 by Ganoi and Mechoulam (Gaoni and Mechoulam
1964), while CBD is the major non-psychotropic cannabinoid found in cannabis.
The other noncannabinoid constituents that have been isolated from cannabis since
2005 belong to 8 major chemical classes, steroids, flavonoids, fatty acids,
428 G. Gupta et al.
13.3.3.3 GW405833
CB receptor, especially cerebrum CB2R, expression, shows dynamic and inducible
profiles under different neurotic conditions. GW405833 sub-atomic compound is
under preclinical investigation showing a particular CB2R agonistic activity. A
recent report revealed that CB2R agonist GW405833 secures liver cells and shows
therapeutic impact by lessening serum aminotransferase levels, and diminishes
hepatocyte apoptosis against intense concanavalin A-initiated poisonous quality
through the CB2 receptors communicated in liver resistant cells (Huang et al. 2019).
Rimonabant was never endorsed in the United States for the treatment of obesity.
The marketing endorsement for the drug was canceled by the European Regulatory
Authorities in 2009 (Sam et al. 2011; Moreira and Crippa 2009).
13.3.5.1 Pain
Therapies based on agonists targeting CB2 receptors have been proposed for the
management of an array of painful conditions. Such conditions include acute pain,
nociceptive, neuropathic pain, and chronic inflammatory pain (Whiteside et al.
2007). CB2 agonists exerted analgesic effects in animal models of neuropathic
pain such as partial sciatic nerve ligation model, spinal nerve ligation model and
chemotherapy-induced neuropathy. In addition, cannabinoids also showed analgesic
effects in diverse persistent inflammatory pain models such as carrageenan, capsai-
cin, complete Freund’s adjuvant, formalin and arachidonic acid (Guindon and
Hohmann 2008). The mechanisms through which cannabinoids alleviate pain
involve decreasing the sensitivity of transient receptor potential channel vanilloid
1 (TRPV1) to noxious stimuli (Jeske et al. 2006), inhibiting NF-κB activity and
microglial production of IL-1β, IL-6 and TNFα (Klegeris et al. 2003).
In clinical trials, there were controversial findings regarding the effect of
cannabinoids in pain management, since some qualitative systematic reviews
reported that cannabinoids are no more effective than codeine in pain management
and the risks associated with their use outweigh their benefit because of their
depressant effects (Campbell et al., 2001).
Table 13.2 Summary of certain diseases that could be targeted by cannabinoids or their
derivatives
Biological Disease Targets Therapeutic Potentials
Pain • Decrease sensitivity of TRPV1 to Acute pain, nociceptive,
noxious stimuli neuropathic and chronic
• Inhibit NF-κB activity and inflammatory pain
microglial production of IL-1β,
IL-6 and TNFα
Metabolic • CB2R blockade Insulin resistance associated with
disorders obesity and obesity-associated
fatty liver
Asthma • Inhibitory effects on mast cells Immunosuppressive, anti-
and eosinophils in lung tissue inflammatory and bronchodilator
• Reduce levels of cytokines effects
involved in the immune response to
an allergen
Glaucoma • Neuroprotective and vasorelaxant Decrease intraocular pressure, and
properties via CB2R activation, hence decrease optic nerve damage
increased aqueous humor outflow due to inadequate blood supply
via enhancing the p42/44 MAP
kinase
Autoimmune • Enhance levels of anti- • Maintain normoglycemia
diseases inflammatory mediators while • Decrease inflammation
decreasing the levels of associated with rheumatoid
pro-inflammatory cytokines arthritis
• Decrease neurodegeneration in
multiple sclerosis
• Improve the symptoms of
Crohn’s, multiple sclerosis
Bone diseases • Stimulation of the CB2R • Prevent osteoclast formation
• Decrease arthritis progression
• Enhance fracture healing
Cardiovascular • CB2R activation • Diminish infract size
disorders
Gastrointestinal • Targeting the CB1 and CB2 • GastricB58 ulcers
disorders receptors • Gastroesophageal reflux
• Irritable bowel syndrome,
• Secretory diarrhea,
• Crohn’s disease,
• Paralytic ileus
• Hepatitis C
Mood and anxiety • CB2R stimulation • Bipolar disorders
disorders • Drug abuse
• Post-traumatic stress disorder
Neurodegenerative • Selective targeting of the CB2R • Multiple sclerosis
diseases • Amyotrophic lateral sclerosis
• Parkinson’s disease
• Huntington’s disease
(continued)
432 G. Gupta et al.
1971 in which there was a reported increase in food intake following use of
Cannabis (Hollister 1971). Another study also presented that oral Δ9-THC doses
of up to 15 mg/day stimulated appetite and produced significant weight gain in
advanced cancer patients (Regelson et al. 1976). Later on, a more comprehensive
study demonstrated clearly that smoking Cannabis leads to a substantial increase in
food intake (Foltin et al. 1986). The expression of CB1 receptor, MAGL and FAAH
in the human pancreas was reported (Kim et al. 2011) and it was recognized that CB1
suppresses β-cell proliferation by hindering insulin secretion. As a result, CB1
receptor blockade leads to elevated β-cell mass in diabetic mice and enhanced insulin
sensitivity. Additionally, the contribution of cannabinoids to the pathogenesis of
diabetic neuropathy, retinopathy and nephropathy has been explored (Horváth et al.
2012).
CB2 receptor stimulation enhanced insulin resistance associated with obesity and
obesity-associated fatty liver and was improved in CB2 knock-out mice, suggesting
that CB2 blockade might be beneficial for the treatment of insulin resistance and fatty
liver (Deveaux et al. 2009).
13.3.5.3 Asthma
Several studies postulated that targeting cannabinoid receptors might be promising
for treating patients with asthma. Cannabinoids demonstrated immunosuppressive,
anti-inflammatory and bronchodilatory effects. In vivo models showed that
cannabinoids exerted inhibitory effects on mast cells and eosinophils in lung tissue
(Giannini et al. 2008) and reduced the levels of cytokines involved in the immune
response to an allergen (Vuolo et al. 2015). Moreover, cannabinoids demonstrated
antibacterial effects against Staphylococci and Streptococci in broth (Van Klingeren
and Ten Ham 1976). CB2 receptors also regulate the function of natural killer cells
by inhibiting cytokine production in a murine model of asthma (Ferrini et al. 2017).
In human studies, early studies found that Cannabis smoke, unlike cigarette
smoke, caused bronchodilatation rather than bronchoconstriction and, unlike
13 Pharmacology of Endocannabinoids and Their Receptors 433
opiates, did not cause central respiratory depression (Vachon et al. 1973). Another
study also demonstrated that doses of THC provided by aerosol caused
bronchodilatation as measured by the enhancement of lung function (Hartley et al.
1978).
13.3.5.4 Glaucoma
Recent studies have demonstrated that the neuroprotective and vasorelaxant
properties of cannabinoids might be effective in reducing intraocular pressure
(Tomida et al. 2004). CB2 receptor activation increased aqueous humor outflow by
enhancing the p42/44 MAP kinase activity in cultured porcine trabecular meshwork
cells (Zhong et al. 2005). According to the American Glaucoma Society,
cannabinoids can decrease intraocular pressure briefly, and reduce blood pressure
and hence can decrease optic nerve damage due to inadequate blood supply (Jampel
2009).
CB2 prevents osteoclast formation (Ofek et al. 2006; Karsak et al. 2005). Other
studies involving in vivo models demonstrated that cannabinoids might improve
fracture healing (Kogan et al. 2015) and prevent the progression of arthritis (Malfait
et al. 2000).
13.3.5.12 Cancers
In addition to palliative effects, preclinical studies demonstrated that cannabinoids
exert antitumor, antiproliferative, antiangiogenic or proapoptotic effects both in vitro
and in vivo against several types of cancer. Such cancers include glioblastoma
(Guzman et al. 2006), glioma (Massi et al. 2004), pancreatic cancer (Carracedo
et al. 2006), oral cancer (Whyte et al. 2010), breast cancer (Caffarel et al. 2010),
prostate cancer (De Petrocellis et al. 2013), lung cancer (Preet et al. 2011), blood
cancer (Gustafsson et al. 2006), liver cancer (Knowles et al. 1980), colorectal cancer
(Kogan et al. 2007), thyroid cancer (Portella et al. 2003), ovarian cancer (Afaq et al.
2006), cervical cancer (Lukhele and Motadi 2016), gastric cancer (Park et al. 2011)
and skin cancer (Blázquez et al. 2006).
In breast cancer cells, cannabinoids have shown the ability to down-regulate Id-1
gene expression and increase the generation of reactive oxygen species leading to
induction of apoptosis and autophagy (Śledziński et al. 2018). In addition, the
antitumor effects of cannabinoids, phytocannabinoids, and synthetic and endoge-
nous ligands have been reported in various types of breast cancer including
hormone-dependent and hormone-independent breast cancer cell lines. These
antitumor effects include induction of apoptosis via pro-caspase-3 cleavage to
caspase-3 and cell cycle arrest, inhibition of angiogenesis by the reduction of
pro-angiogenic factors VEGF, inhibiting forskolin-induced cAMP formation, and
activation of RAF1 translocation and MAPK activity (Kisková et al. 2019).
One of the primary advantages of the potential use of cannabinoids in cancer
management is selectivity and lack of cytotoxicity to normal cells (Guzman 2003).
These effects were observed in cultured cells originating from human, mouse and
436 G. Gupta et al.
rodent tumor models. Clinical trials are needed to demonstrate the effectiveness and
safety of cannabinoids in cancer patients.
13.3.6 Conclusion
References
Abood M, Martin B (1992) Neurobiology of marijuana abuse. Trends Pharmacol Sci 13:201–206
Adler BL, Deleo VA (2019) Allergenic ingredients in commercial topical cannabinoid preparations.
J Am Acad Dermatol. https://doi.org/10.1016/j.jaad.2019.03.015
Adriani W, Caprioli A, Granstrem O, Carli M, Laviola G (2003) The spontaneously hypertensive-
rat as an animal model of ADHD: evidence for impulsive and non-impulsive subpopulations.
Neurosci Biobehav Rev 27:639–651
Afaq F, Sarfaraz S, Syed DN, Khan N, Malik A, Bailey HH, Mukhtar H (2006) Cannabinoid
receptors as a target for therapy of ovarian cancer. AACR Meeting, Washington, DC
Akram H, Mokrysz C, Curran HV (2019) What are the psychological effects of using synthetic
cannabinoids? A systematic review. J Psychopharmacol 33:271–283
Alexander SP (2016) Therapeutic potential of cannabis-related drugs. Prog Neuropsychopharmacol
Biol Psychiatry 64:157–166
Alipour A, Patel PB, Shabbir Z, Gabrielson S (2019) Review of the many faces of synthetic
cannabinoid toxicities. Ment Health Clin 9:93–99
Andrade AK, Renda B, Murray JE (2019) Cannabinoids, interoception, and anxiety. Pharmacol
Biochem Behav 180:60–73
Bara A, Manduca A, Bernabeu A, Borsoi M, Serviado M, Lassalle O, Murphy M, Wager-Miller J,
Mackie K, Pelissier-Alicot AL, Trezza V, Manzoni OJ (2018) Sex-dependent effects of in utero
cannabinoid exposure on cortical function. Elife 7
Baron EP (2018) Medicinal properties of cannabinoids, terpenes, and flavonoids in cannabis, and
benefits in migraine, headache, and pain: an update on current evidence and cannabis science.
Headache 58:1139–1186
Battista N, Di Tommaso M, Bari M, Maccarrone M (2012) The endocannabinoid system: an
overview. Front Behav Neurosci 6:9
Bilbao A, Cippitelli A, Bermudez-Silva FJ, Del Arco I, Navarro M, Rodríguez De Fonseca F (2004)
The endocannabinoid system: physiology and pharmacology. Alcohol Alcohol 40:2–14
Bingham B, Jones P, Uveges A, Kotnis S, Lu P, Smith V, Sun SC, Resnick L, Chlenov M, He Y
(2007) Species-specific in vitro pharmacological effects of the cannabinoid receptor 2 (CB2)
selective ligand AM1241 and its resolved enantiomers. Br J Pharmacol 151:1061–1070
Blankman JL, Simon GM, Cravatt BF (2007) A comprehensive profile of brain enzymes that
hydrolyze the endocannabinoid 2-arachidonoylglycerol. Chem Biol 14:1347–1356
438 G. Gupta et al.
Crowley K, De Vries ST, Moreno-Sanz G (2018) Self-reported effectiveness and safety of Trokie
((R)) lozenges: a standardized formulation for the buccal delivery of cannabis extracts. Front
Neurosci 12:564
Curran HV, Hindocha C, Morgan CJA, Shaban N, Das RK, Freeman TP (2019) Which biological
and self-report measures of cannabis use predict cannabis dependency and acute psychotic-like
effects? Psychol Med 49:1574–1580
Cyr C, Arboleda MF, Aggarwal SK, Balneaves LG, Daeninck P, Neron A, Prosk E, Vigano A
(2018) Cannabis in palliative care: current challenges and practical recommendations. Ann
Palliat Med 7:463–477
Da Silva VK, De Freitas BS, Garcia RCL, Monteiro RT, Hallak JE, Zuardi AW, Crippa JAS,
Schroder N (2018) Antiapoptotic effects of cannabidiol in an experimental model of cognitive
decline induced by brain iron overload. Transl Psychiatry 8:176
Dale T, Downs J, Olson H, Bergin AM, Smith S, Leonard H (2019) Cannabis for refractory epilepsy
in children: a review focusing on CDKL5 Deficiency Disorder. Epilepsy Res 151:31–39
Daris B, Tancer Verboten M, Knez Z, Ferk P (2019) Cannabinoids in cancer treatment: therapeutic
potential and legislation. Bosn J Basic Med Sci 19:14–23
De Petrocellis L, Ligresti A, Schiano Moriello A, Iappelli M, Verde R, Stott CG, Cristino L,
Orlando P, Di Marzo V (2013) Non-THC cannabinoids inhibit prostate carcinoma growth
in vitro and in vivo: pro-apoptotic effects and underlying mechanisms. Br J Pharmacol
168:79–102
Devane W, Hanus L, Breuer A, Pertwee R, Stevenson L, Griffin G, Gibson D, Mandelbaum A,
Etinger A, Mechoulam R (1992) Isolation and structure of a brain constituent that binds to the
cannabinoid receptor. Science 258:1946–1949
Deveaux V, Cadoudal T, Ichigotani Y, Teixeira-Clerc F, Louvet A, Manin S, Tran-Van Nhieu J,
Belot MP, Zimmer A, Even P (2009) Cannabinoid CB2 receptor potentiates obesity-associated
inflammation, insulin resistance and hepatic steatosis. PLoS One 4:e5844
Diao X, Huestis MA (2019) New synthetic cannabinoids metabolism and strategies to best identify
optimal marker metabolites. Front Chem 7:109
Dinis-Oliveira RJ (2019) [The clinical toxicology perspective on the therapeutic use of cannabis and
cannabinoids]. Acta Med Port 32: 87–90
Dol-Gleizes F, Paumelle R, Visentin V, Mares AM, Desitter P, Hennuyer N, Gilde A, Staels B,
Schaeffer P, Bono F (2009) Rimonabant, a selective cannabinoid CB1 receptor antagonist,
inhibits atherosclerosis in LDL receptor-deficient mice. Arterioscler Thromb Vasc Biol
29:12–18
Elliott J, McCoy B, Clifford T, Potter BK, Skidmore B, Wells GA, Coyle D (2019) Cost-
effectiveness of cannabinoids for pediatric drug-resistant epilepsy: protocol for a systematic
review of economic evaluations. Syst Rev 8:75
Ferrini M, Hong S, Stierle A, Stierle D, Stella N, Roberts K, Jaffar Z (2017) CB 2 receptors regulate
natural killer cells that limit allergic airway inflammation in a murine model of asthma. Allergy
72:937–947
Fiore D, Ramesh P, Proto MC, Piscopo C, Franceschelli S, Anzelmo S, Medema JP, Bifulco M,
Gazzerro P (2018) Rimonabant kills colon cancer stem cells without inducing toxicity in normal
colon organoids. Front Pharmacol 8:949
Foltin RW, Brady JV, Fischman MW (1986) Behavioral analysis of marijuana effects on food
intake in humans. Pharmacol Biochem Behav 25:577–582
Fong TM, Guan X-M, Marsh DJ, Shen C-P, Stribling DS, Rosko KM, Lao J, Yu H, Feng Y, Xiao
JC, Van Der Ploeg LHT, Goulet MT, Hagmann WK, Lin LS, Lanza TJ, Jewell JP, Liu P, Shah
SK, Qi H, Tong X, Wang J, Xu SS, Francis B, Strack AM, Macintyre DE, Shearman LP (2007)
Antiobesity efficacy of a novel cannabinoid-1 receptor inverse agonist, N-[(1S,2S)-3-
(4-chlorophenyl)-2-(3-cyanophenyl)-1-methylpropyl]-2-methyl-2-[[5-(trifluoromethyl)pyridin-
2-yl]oxy]propanamide (MK-0364), in rodents. J Pharmacol Exp Ther 321:1013–1022
Fowler C (2007) The contribution of cyclooxygenase-2 to endocannabinoid metabolism and action.
Br J Pharmacol 152:594–601
440 G. Gupta et al.
Friedman D, French JA, Maccarrone M (2019) Safety, efficacy, and mechanisms of action of
cannabinoids in neurological disorders. Lancet Neurol 18:504–512
Galiègue S, Mary S, Marchand J, Dussossoy D, CarriÈre D, Carayon P, Bouaboula M, Shire D,
Fur G, Casellas P (1995) Expression of central and peripheral cannabinoid receptors in human
immune tissues and leukocyte subpopulations. Eur J Biochem 232:54–61
Galiegue S, Mary S, Marchand J, Dussossoy D, Carriere D, Carayon P, Bouaboula M, Shire D, Le
Fur G, Casellas P (1995) Expression of central and peripheral cannabinoid receptors in human
immune tissues and leukocyte subpopulations. Eur J Biochem 232:54–61
Gaoni Y, Mechoulam R (1964) Isolation, structure, and partial synthesis of an active constituent of
hashish. J Am Chem Soc 86:1646–1647
Gertsch J, Pertwee RG, Di Marzo V (2010) Phytocannabinoids beyond the Cannabis plant - do they
exist? Br J Pharmacol 160:523–529
Giang DK, Cravatt BF (1997) Molecular characterization of human and mouse fatty acid amide
hydrolases. Proc Natl Acad Sci U S A 94:2238–2242
Giannini L, Nistri S, Mastroianni R, Cinci L, Vannacci A, Mariottini C, Passani M, Mannaioni P,
Bani D, Masini E (2008) Activation of cannabinoid receptors prevents antigen-induced asthma-
like reaction in guinea pigs. J Cell Mol Med 12:2381–2394
Gobbi G, Bambico FR, Mangieri R, Bortolato M, Campolongo P, Solinas M, Cassano T, Morgese
MG, Debonnel G, Duranti A, Tontini A, Tarzia G, Mor M, Trezza V, Goldberg SR, Cuomo V,
Piomelli D (2005) Antidepressant-like activity and modulation of brain monoaminergic trans-
mission by blockade of anandamide hydrolysis. Proc Natl Acad Sci U S A 102:18620–18625
Goncalves J, Rosado T, Soares S, Simao AY, Caramelo D, Luis A, Fernandez N, Barroso M,
Gallardo E, Duarte AP (2019) Cannabis and its secondary metabolites: their use as therapeutic
drugs, toxicological aspects, and analytical determination. Medicines (Basel) 6. https://doi.org/
10.3390/medicines6010031
Gonsiorek W, Lunn C, Fan X, Narula S, Lundell D, Hipkin RW (2000) Endocannabinoid
2-arachidonyl glycerol is a full agonist through human type 2 cannabinoid receptor: antagonism
by anandamide. Mol Pharmacol 57:1045–1050
Griffin G, Tao Q, Abood ME (2000) Cloning and pharmacological characterization of the rat CB
(2) cannabinoid receptor. J Pharmacol Exp Ther 292:886–894
Guindon J, Hohmann A (2008) Cannabinoid CB2 receptors: a therapeutic target for the treatment of
inflammatory and neuropathic pain. Br J Pharmacol 153:319–334
Gustafsson K, Christensson B, Sander B, Flygare J (2006) Cannabinoid receptor-mediated apopto-
sis induced by R (+)-methanandamide and Win55, 212-2 is associated with ceramide accumu-
lation and p38 activation in mantle cell lymphoma. Mol Pharmacol 70:1612–1620
Guzman M (2003) Cannabinoids: potential anticancer agents. Nature reviews cancer 3:745
Guzman M, Duarte M, Blazquez C, Ravina J, Rosa M, Galve-Roperh I, Sanchez C, Velasco G,
Gonzalez-Feria L (2006) A pilot clinical study of Δ9-tetrahydrocannabinol in patients with
recurrent glioblastoma multiforme. Br J Cancer 95:197–203
Hartley J, Nogrady S, Seaton A (1978) Bronchodilator effect of delta1-tetrahydrocannabinol. Br J
Clin Pharmacol 5:523–525
He Y, De Witte LD, Schubart CD, Van Gastel WA, Koeleman BPC, De Jong S, Ophoff RA, Hol
EM, Boks MP (2019) Liprin alfa 2 gene expression is increased by cannabis use and associated
with neuropsychological function. Eur Neuropsychopharmacol 29:643–652
Henschke P (2019) Cannabis: an ancient friend or foe? What works and doesn’t work. Semin Fetal
Neonatal Med 24:149–154
Herkenham M, Lynn AB, Johnson MR, Melvin LS, De Costa BR, Rice KC (1991) Characterization
and localization of cannabinoid receptors in rat brain: a quantitative in vitro autoradiographic
study. J Neurosci 11:563–583
Ho C, Martinusen D, Lo C (2019) A review of cannabis in chronic kidney disease symptom
management. Can J Kidney Health Dis 6:2054358119828391
Hollander PA, Amod A, Litwak LE, Chaudhari U (2010) Effect of rimonabant on glycemic control
in insulin-treated type 2 diabetes: the ARPEGGIO trial. Diabetes Care 33:605–607
13 Pharmacology of Endocannabinoids and Their Receptors 441
Hollister LE (1971) Hunger and appetite after single doses of marihuana, alcohol, and dextroam-
phetamine. Clin Pharmacol Ther 12:44–49
Holt S, Comelli F, Costa B, Fowler CJ (2005) Inhibitors of fatty acid amide hydrolase reduce
carrageenan-induced hind paw inflammation in pentobarbital-treated mice: comparison with
indomethacin and possible involvement of cannabinoid receptors. Br J Pharmacol 146:467–476
Horváth B, Mukhopadhyay P, Haskó G, Pacher P (2012) The endocannabinoid system and plant-
derived cannabinoids in diabetes and diabetic complications. Am J Pathol 180:432–442
Howlett AC, Barth F, Bonner TI, Cabral G, Casellas P, Devane WA, Felder CC, Herkenham M,
Mackie K, Martin BR, Mechoulam R, Pertwee RG (2002) International Union of Pharmacol-
ogy. Xxvii. Classification of cannabinoid receptors. Pharmacol Rev 54:161–202
Huang Z-B, Zheng Y-X, Li N, Cai S-L, Huang Y, Wang J, Hu X-W, Wang Y, Wu J, Fan X-G
(2019) Protective effects of specific cannabinoid receptor 2 agonist GW405833 on concanavalin
A-induced acute liver injury in mice. Acta Pharmacol Sinica
Huestis MA, Blount BC, Milan DF, Newmeyer MN, Schroeder J, Smith ML (2019) Correlation of
creatinine- and specific gravity-normalized free and glucuronidated urine cannabinoid
concentrations following smoked, vaporized, and oral cannabis in frequent and occasional
cannabis users. Drug Test Anal. https://doi.org/10.1002/dta.2576
Izzo AA, Camilleri M (2008) Emerging role of cannabinoids in gastrointestinal and liver diseases:
basic and clinical aspects. Gut 57:1140–1155
Jampel H (2009) Position statement on marijuana and the treatment of glaucoma. American
Glaucoma Society, San Francisco
Javed H, Azimullah S, Haque ME, Ojha SK (2016) Cannabinoid type 2 (CB2) receptors activation
protects against oxidative stress and neuroinflammation associated dopaminergic
neurodegeneration in rotenone model of Parkinson’s Disease. Front Neurosci 10:321
Jeske NA, Patwardhan AM, Gamper N, Price TJ, Akopian AN, Hargreaves KM (2006) Cannabi-
noid WIN 55,212-2 regulates TRPV1 phosphorylation in sensory neurons. J Biol Chem
281:32879–32890
Jordan CJ, Xi ZX (2019) Progress in brain cannabinoid CB2 receptor research: from genes to
behavior. Neurosci Biobehav Rev 98:208–220
Kaminski NE (1998) Inhibition of the cAMP signaling cascade via cannabinoid receptors: a
putative mechanism of immune modulation by cannabinoid compounds. Toxicol Lett
102:59–63
Karsak M, Cohen-Solal M, Freudenberg J, Ostertag A, Morieux C, Kornak U, Essig J, Erxlebe E,
Bab I, Kubisch C (2005) Cannabinoid receptor type 2 gene is associated with human osteopo-
rosis. Human Mol Genet 14:3389–3396
Kathuria S, Gaetani S, Fegley D, ValiñO F, Duranti A, Tontini A, Mor M, Tarzia G, Rana GL,
Calignano A, Giustino A, Tattoli M, Palmery M, Cuomo V, Piomelli D (2002) Modulation of
anxiety through blockade of anandamide hydrolysis. Nat Med 9:76
Kaur R, Singh P, Kaur A, Mahajan D, Kaur H (2013) Effect of rimonabant on the components of
metabolic syndrome: a randomized, controlled study done on Punjabi population. J Pharm
Negat Results 4:46–53
Khuja I, Yekhtin Z, Or R, Almogi-Hazan O (2019) Cannabinoids reduce inflammation but inhibit
lymphocyte recovery in murine models of bone marrow transplantation. Int J Mol Sci 20. https://
doi.org/10.3390/ijms20030668
Kim W, Doyle ME, Liu Z, Lao Q, Shin Y-K, Carlson OD, Kim HS, Thomas S, Napora JK, Lee EK
(2011) Cannabinoids inhibit insulin receptor signaling in pancreatic β-cells. Diabetes
60:1198–1209
Kisková T, Mungenast F, Suváková M, Jäger W, Thalhammer T (2019) Future aspects for
cannabinoids in breast cancer therapy. Int J Mol Sci 20:1673
Klegeris A, Bissonnette CJ, Mcgeer PL (2003) Reduction of human monocytic cell neurotoxicity
and cytokine secretion by ligands of the cannabinoid-type CB2 receptor. Br J Pharmacol
139:775–786
Knowles BB, Howe CC, Aden DP (1980) Human hepatocellular carcinoma cell lines secrete the
major plasma proteins and hepatitis B surface antigen. Science 209:497–499
442 G. Gupta et al.
Koch M, Varela L, Kim JG, Kim JD, Hernández-NuñO F, Simonds SE, Castorena CM, Vianna CR,
Elmquist JK, Morozov YM (2015) Hypothalamic POMC neurons promote cannabinoid-
induced feeding. Nature 519:45–50
Kogan NM, Schlesinger M, Peters M, Marincheva G, Beeri R, Mechoulam R (2007) A cannabinoid
anticancer quinone, HU-331, is more potent and less cardiotoxic than doxorubicin: a compara-
tive in vivo study. J Pharmacol Exp Ther 322:646–653
Kogan NM, Melamed E, Wasserman E, Raphael B, Breuer A, Stok KS, Sondergaard R, Escudero
AV, Baraghithy S, Attar-Namdar M (2015) Cannabidiol, a major non-psychotropic cannabis
constituent enhances fracture healing and stimulates lysyl hydroxylase activity in osteoblasts. J
Bone Miner Res 30:1905–1913
Korem N, Akirav I (2014) Cannabinoids prevent the effects of a footshock followed by situational
reminders on emotional processing. Neuropsychopharmacology 39:2709–2722
Kowal M, Hazekamp A, Grotenhermen F (2016) Review on clinical studies with cannabis and
cannabinoids 2010-2014
Krebs MO, Kebir O, Jay TM (2019) Exposure to cannabinoids can lead to persistent cognitive and
psychiatric disorders. Eur J Pain. https://doi.org/10.1002/ejp.1377
Kumar A, Premoli M, Aria F, Bonini SA, Maccarinelli G, Gianoncelli A, Memo M, Mastinu A
(2019) Cannabimimetic plants: are they new cannabinoidergic modulators? Planta
249:1681–1694
Lattanzi S, Trinka E, Russo E, Striano P, Citraro R, Silvestrini M, Brigo F (2019) Cannabidiol as
adjunctive treatment of seizures associated with Lennox-Gastaut syndrome and Dravet syn-
drome. Drugs Today (Barc) 55:177–196
Li X, Kaminski NE, Fischer LJ (2001) Examination of the immunosuppressive effect of Δ
9-tetrahydrocannabinol in streptozotocin-induced autoimmune diabetes. Int Immunopharmacol
1:699–712
Li Q, Wang F, Zhang Y-M, Zhou J-J, Zhang Y (2013) Activation of cannabinoid type 2 receptor by
JWH133 protects heart against ischemia/reperfusion-induced apoptosis. Cell Physiol Biochem
31:693–702
Liu J, Wang L, Harvey-White J, Huang BX, Kim H-Y, Luquet S, Palmiter RD, Krystal G, Rai R,
Mahadevan A (2008) Multiple pathways involved in the biosynthesis of anandamide. Neuro-
pharmacology 54:1–7
Lopez-Rodriguez AB, Siopi E, Finn DP, Marchand-Leroux C, Garcia-Segura LM, Jafarian-
Tehrani M, Viveros M-P (2013) CB1 and CB2 cannabinoid receptor antagonists prevent
minocycline-induced neuroprotection following traumatic brain injury in mice. Cereb Cortex
25:35–45
Lossignol D (2019) Cannabinoids: a new approach for pain control? Curr Opin Oncol. https://doi.
org/10.1097/CCO.0000000000000523
Lowin T, Schneider M, Pongratz G (2019) Joints for joints: cannabinoids in the treatment of
rheumatoid arthritis. Curr Opin Rheumatol 31:271–278
Lu M, Agito MD (2015) Cannabinoid hyperemesis syndrome: marijuana is both antiemetic and
proemetic. Cleve Clin J Med 82:429–434
Lukhele ST, Motadi LR (2016) Cannabidiol rather than Cannabis sativa extracts inhibit cell growth
and induce apoptosis in cervical cancer cells. BMC Complement Altern Med 16:335
Mackie K, Devane WA, Hille B (1993) Anandamide, an endogenous cannabinoid, inhibits calcium
currents as a partial agonist in N18 neuroblastoma cells. Mol Pharmacol 44:498–503
Malfait A, Gallily R, Sumariwalla P, Malik A, Andreakos E, Mechoulam R, Feldmann M (2000)
The nonpsychoactive cannabis constituent cannabidiol is an oral anti-arthritic therapeutic in
murine collagen-induced arthritis. Proc Natl Acad Sci U S A 97:9561–9566
Massi P, Vaccani A, Ceruti S, Colombo A, Abbracchio MP, Parolaro D (2004) Antitumor effects of
cannabidiol, a nonpsychoactive cannabinoid, on human glioma cell lines. J Pharmacol Exp Ther
308:838–845
Matsuda LA, Lolait SJ, Brownstein MJ, Young AC, Bronner TI (1990) Structure of a cannabinoid
receptor and functional expression of the cloned cDNA. Nature 346:561
13 Pharmacology of Endocannabinoids and Their Receptors 443
May MB, Glode AE (2016) Dronabinol for chemotherapy-induced nausea and vomiting unrespon-
sive to antiemetics. Cancer Manag Res 8:49–55
Mechoulam R, Ben-Shabat S, Hanus L, Ligumsky M, Kaminski NE, Schatz AR, Gopher A,
Almog S, Martin BR, Compton DR (1995) Identification of an endogenous 2-monoglyceride,
present in canine gut, that binds to cannabinoid receptors. Biochem Pharmacol 50:83–90
Miller LK, Devi LA (2011) The highs and lows of cannabinoid receptor expression in disease:
mechanisms and their therapeutic implications. Pharmacol Rev 63:461–470
Moreira FA, Crippa JAS (2009) The psychiatric side-effects of rimonabant. Braz J Psychiatry
31:145–153
Mukherjee S, Adams M, Whiteaker K, Daza A, Kage K, Cassar S, Meyer M, Yao BB (2004)
Species comparison and pharmacological characterization of rat and human CB 2 cannabinoid
receptors. Eur J Pharmacol 505:1–9
Munro S, Thomas KL, Abu-Shaar M (1993) Molecular characterization of a peripheral receptor for
cannabinoids. Nature 365:61
Naftali T, Mechulam R, Lev LB, Konikoff FM (2014) Cannabis for inflammatory bowel disease.
Dig Dis 32:468–474
Nagarkatti P, Pandey R, Rieder SA, Hegde VL, Nagarkatti M (2009) Cannabinoids as novel anti-
inflammatory drugs. Future Med Chem 1:1333–1349
National Research Council (US) Committee on Substance Abuse and Habitual Behavior 1982) An
analysis of Marijuana Policy [Online]. Accessed https://www.ncbi.nlm.nih.gov/books/
NBK217602/
Navarrete F, Pérez-Ortiz JM, Manzanares J (2012) Cannabinoid CB2 receptor-mediated regulation
of impulsive-like behaviour in DBA/2 mice. Br J Pharmacol 165:260–273
Nunez E, Benito C, Pazos MR, Barbachano A, Fajardo O, Gonzalez S, Tolon RM, Romero J (2004)
Cannabinoid CB2 receptors are expressed by perivascular microglial cells in the human brain:
an immunohistochemical study. Synapse 53:208–213
Ofek O, Karsak M, Leclerc N, Fogel M, Frenkel B, Wright K, Tam J, Attar-Namdar M, Kram V,
Shohami E (2006) Peripheral cannabinoid receptor, CB2, regulates bone mass. Proc Natl Acad
Sci U S A:103, 696–701
Okamoto Y, Morishita J, Tsuboi K, Tonai T, Ueda N (2004) Molecular characterization of a
phospholipase D generating anandamide and its congeners. J Biol Chem 279:5298–5305
Onaivi ES, Ishiguro H, Gong JP, Patel S, Perchuk A, Meozzi PA, Myers L, Mora Z, Tagliaferro P,
Gardner E, Brusco A, Akinshola BE, Liu QR, Hope B, Iwasaki S, Arinami T, Teasenfitz L, Uhl
GR (2006) Discovery of the presence and functional expression of cannabinoid CB2 receptors
in brain. Ann N Y Acad Sci 1074:514–536
Onaivi ES, Ishiguro H, Gong JP, Patel S, Meozzi PA, Myers L, Perchuk A, Mora Z, Tagliaferro PA,
Gardner E, Brusco A, Akinshola BE, Hope B, Lujilde J, Inada T, Iwasaki S, Macharia D,
Teasenfitz L, Arinami T, Uhl GR (2008) Brain neuronal CB2 cannabinoid receptors in drug
abuse and depression: from mice to human subjects. PLoS One 3:e1640
Park JM, Xian XS, Choi MG, Park H, Cho YK, Lee IS, Kim SW, Chung IS (2011) Antiproliferative
mechanism of a cannabinoid agonist by cell cycle arrest in human gastric cancer cells. J Cell
Biochem 112:1192–1205
Pertwee R (1997) Pharmacology of cannabinoid CB1 and CB2 receptors. Pharmacol Ther
74:129–180
Pertwee RG (2005) The therapeutic potential of drugs that target cannabinoid receptors or modulate
the tissue levels or actions of endocannabinoids. AAPS J 7
Pertwee R (2010) Receptors and channels targeted by synthetic cannabinoid receptor agonists and
antagonists. Curr Med Chem 17:1360
Pertwee R, Howlett A, Abood ME, Alexander S, Di Marzo V, Elphick M, Greasley P, Hansen HS,
Kunos G, Mackie K (2010) International Union of Basic and Clinical Pharmacology. LXXIX.
Cannabinoid receptors and their ligands: beyond CB1 and CB2. Pharmacol Rev 62:588–631
Piomelli D (2003) The molecular logic of endocannabinoid signalling. Nat Rev Neurosci
4:873–884
444 G. Gupta et al.
Abstract
Keywords
Abbreviations
AD Androstanediol
AMPA α-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid
ANT Adenine nucleotide transporter protein
ARH Arcuate nucleus of the hypothalamus
BLSA Baltimore Longitudinal Study of Aging
CB1 Type-1 cannabinoid
CNS Central nervous system
CREB cAMP response element binding protein
D1 Dopamine 1
D2 Dopamine 2
DHEA Dehydroepiandrosterone
DHP Dihydroprogesterone
ERɑ Estrogen receptor ɑ
ERs Estrogen receptors
ERβ Estrogen receptor β
ET Essential tremor
GABA Gamma-aminobutyric acid
GAMSA GABAA receptor modulating steroid antagonists
GPER G protein-coupled estrogen receptor
GPER1 G protein-coupled estrogen receptor 1
GSK3b Glycogen synthase kinase 3b
HPA Hypothalamic-pituitary-adrenal
IP3 Inositol 1,4,5-trisphoshate
LXRs Liver X receptors
MAPK Mitogen-activated protein kinase
MDD Major depressive disorder
mERɑ Membrane-associated estrogen receptor ɑ
mERs Membrane-associated estrogen receptors
mERβ Membrane-associated estrogen receptor β
mGluR Metabotropic glutamate receptor
MPN Medial preoptic nucleus
mPRs Membrane-bound progesterone receptors
MPTP 1-Methyl 4-phenyl-1,2,3,6 tetrahydropyridine
NADPH Nicotinamide adenine dinucleotide phosphate
NMDA N-methyl-D-aspartate
P4507ɑ Cytochrome P450 7ɑ-hydroxylase
14 Hormones and Steroids as Neurotransmitters 449
14.1 Introduction
Neurons in the brain signal to their neighbors via the aid of endogenous chemicals
called neurotransmitters. Neurotransmitters are chemical messengers synthesized by
neurons, which enable interconnection of nerve fibers within their vicinity. In a
broader sense, neurotransmitters are chemical messengers which communicate
signals between chemical synapses including neuromuscular junctions, from one
neuron to another "target" neuron, muscle cells or gland cells (Badgaiyan 2011). Till
the very recent past, neurotransmitters belonging to the categories of amino acid and
their derivatives, chains of amino acids, peptides or proteins was a very stated fact
(Rudolph et al. 2016). Molecules such as steroids, cholesterol derivatives and
hormones were never believed to be involved in the neurotransmission system
since they easily pass through cell membranes and distribute themselves in tissue.
It was found that steroids exert an acute effect on physiology (Szego and Davis
1967), neuron activity (Kelly et al. 1976) and the expression of behavior (Hayden-
450 S. Marwein et al.
Hixson and Ferris 1991) that can happen within minutes. Their ability to modulate
the neuronal function within minutes and to rapidly stimulate cognitive functions
and behaviors suggest their neurotransmitter-like action. The understanding of the
role of neuroactive steroids was conceived from the study by Etienne-Emile Baulieu
on the levels of dehydroepiandrosterone (DHEA) sulfate in the brain of adult male
rats (Corpechot et al. 1981; Baulieu 1998). In another study, Corpéchot and his
group attained a significant finding which describes the endogenous production of
DHEA sulfate within the brain, without considering the steroids secreted from the
adrenals and the gonads (Corpechot et al. 1981). The steroids produced within the
nervous system were referred to as neurosteroids (Corpechot et al. 1981). The
documentation of other classes of steroid receptor such as the pregnane X receptor
(PXR) that can be stimulated by steroids like pregnenolone and progesterone
(Kliewer et al. 1998; Moore et al. 2000) additionally augments to the element of
brain action modulating activity of steroids. From the 1970s onward, the potential
effects of steroids such as estradiol, a cholesterol derivative, in the nervous system
were appreciated owing to their ability to cause rapid alteration of the electrical
impulse fired by specific neurons within the brain. This rapid change and the
response time are not achievable by the binding of estrogen to the nuclear steroid
receptor within the neurons, which is a slow process that takes hours to be initiated
(Majewska et al. 1986; Paul and Purdy 1992). This was the initial evidence of the
possible mechanism that certain steroids and hormones can modulate excitability of
neurons by interacting with specific cell surface neurotransmitter receptors (Lambert
et al. 1995; Rupprecht 1997; Rupprecht and Holsboer 1999). The modulatory effects
of steroids in between the neurons occurs rapidly, ranging from milliseconds to
seconds, which is distinguishable from the steroid’s action at the genome, since it
involves an event ranging from minutes to hours depending on the proportion of
protein biosynthesis (McEwen 1991). Therefore, the activity of steroids within the
brain that can be either their genomic or non-genomic effects establishes the
understanding at the molecular level of a comprehensive effect of steroids on
neuronal excitability and plasticity. Several studies demonstrate that steroids can
and do function in ways that are “neurotransmitter-like,” as they are produced locally
in a specific region within neural circuits whereby they can exert their modulating
activity locally within minutes to stimulate neuronal functions such as cognition and
behavior-related activity (Balthazart et al. 2006; Dewing et al. 2007; Saldanha et al.
2011; Remage-Healey 2014).
Thus, steroids which have the capability of binding to the neurotransmitter
receptors and modulate the neurotransmission signal are included together within
the term neurosteroids or neuroactive steroids. However, neurosteroids are actually
metabolic products of cholesterol which are produced continuously inside the brain
from the readily available metabolic precursors and enzymes and they initiate instant
effects on neuronal excitability (Carver and Reddy 2013). Neuroactive steroids are
steroids synthesized in the endocrine gland that circulates through the bloodstream
into the brain to exert their effects on brain function. Therefore, neuroactive steroids
include neurosteroids and other steroids that are produced within the adrenal, testis
14 Hormones and Steroids as Neurotransmitters 451
Estradiol
Neuroactive steroid Progesterone
Glucocorticoid
Pregnenolone
Progesterone
Allopregnenolone
Dehydroepiandrosterone
Dehydroepiandrosterone
sulfate
and ovary, and are transported into the brain through the blood-brain barrier to
stimulate brain action in a similar way to the neurosteroids (Girdler et al. 2012).
Neuroactive steroids have the ability to regulate neuronal excitability function
and are of different categories depending on their source and site of production
(Zheng 2009). Neuroactive steroids are divided into two categories on the basis of
the source and production site, that is, the exogenous steroids (these include the
synthetic steroids) and endogenous steroids (Fig. 14.1). Further, based on the site of
production, endogenous steroids are subdivided into hormonal steroid (steroids
synthesized within the endocrine glands) and neurosteroids (steroids synthesized
within the brain) (Melcangi et al. 2008). The examples of hormonal steroids mostly
consist of steroids synthesized in the ovary such as progesterone and estradiol, in the
testis such as testosterone, and in the adrenal gland including DHEA and glucocor-
ticoid (Rhodes et al. 2004; Scharfman and MacLusky 2006), whereas the
neurosteroids include steroids produced by the neuronal and glial cells, with
DHEA, pregnenolone, allopregnanolone, progesterone, and their sulfate esters as
the well-known examples (Baulieu 1998). Additionally, several synthetic steroids
such as alphaxalone and steroid-3ɑ-hydroxy-5β-pregnan-20-one hemisuccinate pos-
sess similar characteristics of modulating neuronal activities to the endogenous
steroids (Melcangi et al. 2008). Neuroactive steroids also include
allotetrahydrodeoxycorticosterone (THDOC), and androstanediol (AD),
ganaloxone, androsterone, etiocholanolone (Akk et al. 2005, 2007), and 17-α and
17-β estradiol (Nguyen et al. 2017).
452 S. Marwein et al.
The synthesis of neuroactive steroids occurs in the brain, the gonads, the adrenal
glands and even in the fetoplacental unit within the body (Midzak et al. 2011). In
peripheral tissues, the gonads and adrenal gland are the main site of neuroactive
steroid production. Further, the metabolization process into active steroid
metabolites occurs in tissues including the endocrine tissues and liver (Akk et al.
2009). The synthesis of neuroactive steroids within the body is catalyzed with the aid
of various enzymes. For instance, the steroid 5α-reductase, an NADPH-dependent
enzyme, catalyzes the reduction of testosterone to the neuroactive steroid, dihydro-
testosterone (Reddy and Rogawski 2010). These neuroactive steroids are then
circulated into the bloodstream, crossing through the blood-brain barrier into the
brain (Haage and Johansson 1999; Baulieu et al. 2001). 5α-Reductase enzyme also
catalyzes the reduction of steroids such as deoxycorticosterone, progesterone and
various 3-keto-pregnane steroids and is available in tissues such as the brain, liver,
prostate, genitalia and testis. The enzyme 5α-reductase exerts its activity in brain
regions such as the neurons, glial cells, neocortex, subcortical white matter and the
hippocampus (Appelgren 1967). Other enzymes necessary for the biosynthesis of
neuroactive steroids include aromatase, sulfotransferase sulfatase, cytochrome 7ɑ-
hydroxylase (P4507ɑ), 11β-hydroxylase (P450C11β), cytochrome P450 17ɑ-
hydroxylase (P450C17), cytochrome P450 21-hydroxylase (P450C21), 3ɑ-
hydroxysteroid dehydrogenase (3ɑ-HSD), 3β-hydroxysteroid dehydrogenase
(3β-HSD), 5ɑ-reductase and 17β-hydroxysteroid dehydrogenase (17β-HSD)
(Do Rego et al. 2009).
Within the nervous system neuroactive steroids are produced locally, mainly
from cholesterol (Ellsworth et al. 1998), in specific brain tissues such as the pineal
gland which is the main neurosteroidogenic organ, glutamatergic neurons, cortex
and the hippocampal region (Appelgren 1967). These sites are steroidogenic sites
and produce steroidogenic enzymes such as 5α-reductase, aromatase,
sulfotransferase sulfatase, cytochrome 7ɑ-hydroxylase, 11β-hydroxylase and many
other enzymes necessary for steroid production. Neuroactive steroids are
synthesized directly from cholesterol via a progressive A-ring reduction process
(Melcangi et al. 2008). The primary stage in steroid synthesis is the metabolization
of cholesterol into pregnenolone. This step involves the transportation of cholesterol
from the outer to the inner mitochondrial membrane with the aid of molecular
complex comprising proteins such as the translocator protein of 18 kDa (TSPO),
the adenine nucleotide transporter protein (ANT), the steroidogenic acute regulatory
protein (StAR), and the voltage-dependent anion channel protein (VDAC)
(Morohaku et al. 2014; Papadopoulos et al. 2018; Selvaraj and Stocco 2015; Selvaraj
et al. 2015). It is located inside the inner mitochondrial membrane, where cholesterol
is metabolized into pregnenolone in the presence of the steroidogenic enzyme
P450scc (Midzak et al. 2011). Pregnenolone then undergoes transformation to
other steroids via sequent stages inside the endoplasmic reticulum. For instance,
pregnenolone when catalyzed by 3β-HSD enzyme or cytochrome P450C17 is
14 Hormones and Steroids as Neurotransmitters 453
Primarily, steroids and hormones were conceived to act solely through the traditional
genomic pathway by binding to the known steroid receptors (Evans 1988; Paul and
Purdy 1992). However, the documentation of new binding sites for neuroactive
steroids such as progesterone, testosterone (Ramirez and Zheng 1996),
glucocorticoids (Orchinik et al. 1991), estradiol (Pappas et al. 1995) or aldosterone
(Wehling 1997) in the membrane of cells and tissues with different signal transduc-
tion pathways that differ from the conventional transduction pathway involved in
steroid action (Wehling 1997) has shed new light on the primarily known concept.
The neuroactive steroids exert several physiological activities within the brain
facilitated by their interaction with the nuclear/membrane steroid receptors (estrogen
and progesterone receptors), androgen receptors and glucocorticoid receptors and
through their modulating activity toward the ion channels, which include the
GABAA (gamma-aminobutyric acid), NMDA (N-methyl-D-aspartate), AMPA
(α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) and kainate receptors
(Ogden and Traynelis 2011). The neuroactive steroids also exert their neuronal
modulation function through various voltage-dependent ion channels which include
the T-type Ca2+ channel, Na+ channel, Ca2+ activated K+ channel and anion
454
Dehydroepiandroste Androstenediol
OMM Testosterone
17 -hdroxypregnenolone rone
TSPO
P450
StAR aromatase
Cholesterol 17 -
VDAC Estradiol
hydroxylase
3 -HSD
5 -reductase
5 -dihydroprogesterone
Endoplasmic
reticulum
O
O HO
HO
HO
HO
O H
HST
Corticosterone Allotetradeoxycorticosterone
O STS
HO OH
HO S O
HO 7-alpha hydroxy pregnenolone
O
Cholesterol sulfate Cholesterol
O P450scc
P450
O O
HO HO
O O
HST
O
HO S O STS
O O
Pregnenolone sulfate O
HO O H
Pregnenolone 11-Deoxycorticosterone
Progesterone Dihydrodeoxycorticosterone
O
O O
O O
OH
OH
HO
O H
HO H
HO OH O
Dihydroprogesterone Tetrahydroprogesterone
7-alpha hydroxy DHEA 17-hydroxy pregnenolone 17-Hydroxy progesterone
O
O
O
O
HO S O
O
O HO
DHEA sulfate Androstenedione
Dehydroepiandrosterone
(DHEA)
O OH
HO O
Testosterone
Estrone OH
OH
OH
HO
O
H
HO
Androstanediol
Estradiol Dihydrotestosterone
Fig. 14.3 Biochemical pathways for the synthesis of neuroactive steroids in the brain. AROM,
aromatase; HST, sulfotransferase; STS, sulfatase; P450scc, cytochrome P450 side-chain cleavage;
P4507ɑ, cytochrome 7ɑ-hydrolase; P450c11β, 11β-hydrolase; P450C17, cytochrome P450 17ɑ-
-hydrolase/C17,20-lyase; P450C21, 21-hydrolase; 3ɑ-HSD, 3ɑ-hydroxysteroid dehydrogenase;
3β-HSD, 3β-hydroxysteroid dehydrogenase; 5ɑ-R, 5ɑ-reductase; 17β-HSD, 17β-hydroxysteroid
dehydrogenase
channels, and the high-voltage activated Ca2+ channel (Carrer et al. 2003; Todorovic
et al. 2004; Pathirathna et al. 2005; Joksovic et al. 2007; Cheng et al. 2008). It is
understood that the physiological effects of neuroactive steroids result from their
interaction with definite receptors. Therefore, it is necessary to study the mechanism
by which these steroids activate these receptors and affect brain physiology and the
impact of the neurotransmitter receptors on neuroactive steroid actions (Rossetti
et al. 2016).
456 S. Marwein et al.
The discovery of estradiol binding sites in different rat tissues by Elwood V. Jensen
in the early 1950s led to the proposition of the occurrence of an explicit receptor for
estradiol (Jensen 1962). Thereafter, Toft and Gorski (1966) isolated an estrogen
receptor from rat uterus, which was later identified as estrogen receptor α (ERα)
shortly after the discovery of the additional estrogen receptor known as estrogen
receptor β (ERβ). These two functionally similar estrogen receptors (ERs) originated
from different genes (Kuiper et al. 1996). Both the ERɑ and ERβ receptors are also
expressed on the cell membrane (Coleman and Smith 2001; Pedram et al. 2007;
Boonyaratanakornkit 2011; Meitzen et al. 2013), but there is still a lack of under-
standing of their cell membrane association. However, it is suggested to be involved
in post-translational lipid modification and communication with membrane/cyto-
plasmic scaffolding proteins (Boonyaratanakornkit 2011; Meitzen et al. 2013;
Pedram et al. 2007). In addition to the membrane-associated mERɑ and mERβ,
one more membrane-associated estrogen receptor is also known to be involved in the
neurotransmission function of the neuroactive steroids, G protein-coupled estrogen
receptor 1 (GPER1) (Carmeci et al. 1997). All these estrogen receptors were found to
be commonly expressed in the neuronal and glial cells within the central nervous
system (Cardona-Gomez et al. 2000; Chaban et al. 2004; Quesada et al. 2007). Brain
regions such as astrocytes were also found to express ERα, ERβ, GPER and
Gq-mER (Kuo et al. 2010; Almey et al. 2012), and all these receptors are activated
by neuroactive steroids such as estradiol and contribute to the regulation of the
astrocytic functions (Pawlak et al. 2005; Kuo et al. 2010; Karki et al. 2014). For
instance, triggering membrane-associated estrogen receptor by neuroactive steroid
can alter membrane permeability (Fu and Simoncini 2008), initiate second messen-
ger cascades (Coleman and Smith 2001) and hyperpolarize neurons in the preoptic
area (Bologa et al. 2006; Prossnitz et al. 2008). Additionally, cumulative data from
different studies indicate that the dihydrotestosterone metabolite, 3β-diol, can inter-
act with and trigger the ERβ, thereby exerting its oestrogenic effects on various
pathophysiological brain functions, including anxiety, depression, affection
disorders and cognition (Fugger et al. 2000; Krȩżel et al. 2001; Österlund et al.
2005; Kudwa et al. 2006; Handa et al. 2008).
The progesterone receptors are important for the neuronal function of steroids such
as progesterone, metabolites of progesterone and sulfated progesterone. Comparable
to the estrogen receptors, progesterone receptors are of two isoforms, that is PR-A
and PR-B, and unlike ERɑ and ERβ originated from an identical gene (Pratt and Toft
2003). Bound progesterone receptors dissociate from the chaperone proteins, and
undertake conformational alteration leading to their dimerization, thus resulting in
the direct interaction with progesterone response elements (PREs) in promoter
regions of targeted genes through their binding with the steroid receptor coactivators
14 Hormones and Steroids as Neurotransmitters 457
Neuroactive glucocorticoids such as corticosterone and cortisol are also known for
their stimulating activity in various brain-related functions and exhibit their effect
mainly through the glucocorticoid receptors. Glucocorticoid receptors are nuclear
receptors located in the cytoplasm that can be included in the group of receptors by
which the neuroactive steroids (glucocorticoids) facilitate their neuronal signaling
activity (Aranda and Pascual 2001). Classical glucocorticoid receptor signaling
requires ligand binding to initiate glucocorticoid receptor dimerization and translo-
cation to the nucleus (Oakley and Cidlowski 2013) where they bind directly with
DNA and other transcription factors to regulate gene transcription. Glucocorticoids
can activate these receptors at the membrane level to alter the physiology functioning
more like neurotransmitters. The enzymes necessary for corticosterone production
are widely distributed throughout the brain (MacKenzie et al. 2000), and de novo
corticosterone synthesis from pregnenolone has been observed in the hippocampus
of adrenalectomized rats (Higo et al. 2011). This suggests the synthesis of
glucocorticoids, the expression of their receptors within the nervous system and
their responsibility in modulating brain function.
The neuroactive steroids usually bind with the steroid receptors to exert their
physiological actions. However, they are also known to modulate various other
ion channels including the voltage-dependent ion channels, especially GABAA,
NMDA and AMPA receptors. There is ample evidence indicating how neuroactive
458 S. Marwein et al.
Presynaptic
terminal
1 β2 4 β2 GABA site
Neurosteroid
site γ2 δ 4
GABA site β2 1 β2
Cl-
Ca2+ Neurosteroids
Neurosteroids GABA Glutamate Na+
Glycine
NR2 NR1
γ
extracellular extracellular NR2 NR1
β β
intracellular intracellular
K+
Fig. 14.5 Schematic illustration of neuroactive steroid actions mediated by GABAA receptor and
NMDA receptor
which results in brain function stimulating activity such as regulation of mood and
learning via their indirect involvement with the T-type voltage-gated Ca2+ channels
(Smith and Woolley 2004; Benarroch 2007; Grassi et al. 2007; Fatehi and Fatehi-
Hassanabad 2008). They have also been demonstrated to modulate mitochondrial
function and synaptic plasticity and to play a vital role in neuroprotective activity
due to their interaction with the voltage-dependent anion channel receptors (Tuem &
Atey 2017). Furthermore, neuroactive steroids are described to have modulating
effects on every synaptic transmission, namely GABAergic, glutamatergic, dopami-
nergic, cholinergic, serotonergic and noradrenergic synaptic transmission, through
their ability to alter the receptivity of postsynaptic receptors or to alter presynaptic
neurotransmitter release (Belelli et al. 2006; Gibbs et al. 2006; Mitsushima et al.
2007; Pérez-Neri et al. 2008; Laconi et al. 2007). Though with less efficacy, the
neuroactive steroids tetrahydroprogesterone and 3α-diol (Carver and Reddy 2013)
indeed stimulate neurotransmission via the GABAA receptor, thereby mediating
their effects on affection and behavior (Belelli and Lambert 2005). Similarly,
sulfate-conjugated neuroactive steroids such as pregnenolone and DHEA also
exert a weak antagonistic effect on the NMDA receptor at the micromolar level,
thereby producing a stimulating neuronal excitatory action (Rupprecht 2003). Neu-
roactive steroids including progesterone can also bind with the membrane receptors
such as the membrane-associated protein PGRMC1, also known as 25-Dx, and the
membrane progesterone receptors (mPRs) (Singh et al. 2013; Giatti et al. 2016). The
recently identified nuclear pregnane-X receptor (PXR) presents an additional site for
neuroactive steroids to bind and exert their actions (Frye 2011).
It is clear that neuroactive steroids exert various types of physiological activity in
the brain (Lapchak and Araujo 2001; Belelli et al. 2006; Strous et al. 2006) either
through interaction with nuclear steroid receptors such as the estrogen receptors,
androgen receptors and progesterone receptors or through interaction with mem-
brane receptors such as membrane progesterone receptors (Fig. 14.6). In particular,
neuroactive steroids interact with various ion channels (Fig. 14.6) and act as
allosteric modulators of the GABAA receptors (Covey et al. 2001; Belelli and
Lambert 2005), NMDA receptors (Mameli et al. 2005; Monnet and Maurice
2006), kainate receptors (Costa et al. 2000), AMPA receptors (Dubrovsky 2005),
sigma receptors (Maurice et al. 2006), serotonin receptors (Shannon et al. 2005a, b),
nicotinic receptors (Paradiso et al. 2000) and muscarinic receptors (Horishita et al.
2005). Additionally, it has been reported that neuroactive steroids may directly
trigger G protein-coupled membrane receptors (Meyer et al. 2002; Schiess and
Partridge 2005) or indirectly control the binding of neuropeptides to their receptors
(Torres and Ortega 2003).
14 Hormones and Steroids as Neurotransmitters 461
Sigma 1
AMPA
mPR
PGRMC1
GABAA
ER receptor
PXR
Nucleus
PR
Alpha 1 AR
adrenergic
L-type
Ca2+
NMDA
D1
Nicotine
receptor
Fig. 14.6 Schematic illustration of different receptors involved in the neuronal function of the
neuroactive steroids
The general down signaling pathway for most of the neuroactive steroids occurs
through a series of processes as depicted in Fig. 14.7. First, neuroactive steroids
might bind to ion channels (i.e., GABAA, NMDA, AMPA, sigma 1, kainite
receptors) associated with neurotransmitter receptors (1). Second, they can bind
and activate the known steroid receptors widely distributed in the plasma membrane
(2), or they may bind to the nuclear plasma membrane-associated steroid receptors
(3) or bind with the traditional cytoplasmic-associated nuclear steroid receptors (4).
462 S. Marwein et al.
1 G proteins, AMPc,
4
IP3, Ca2+, MAPK,
PI3K, Akt, GSK3β
The neuronal activity of estrogens can stimulate various brain-related activities and
behavior. Abundant evidence describing the relationship of estrogens with brain
disorders has been attained through different investigations in female animals or
women. These studies have demonstrated the role of estrogens in brain-related
disorders (Walf and Frye 2006). Women are observed to be susceptible to depression
when there are fluctuations in the concentration of sex hormones, especially estro-
gen, which is believed to be responsible for disorders including post-partum depres-
sion, premenstrual dysphoric disorder, and perimenopausal or postmenopausal
depression (Thorpe et al. 2001). On the other hand, estrogens can display
neuroprotective effects when abundant amounts of endogenous estrogens are pro-
duced within the brain (Carswell et al. 2000; McCullough et al. 2003). Estrogens are
reported to display a vital role in processes such as neuronal plasticity and spine
14 Hormones and Steroids as Neurotransmitters 465
synapse formation (Herrick et al. 2006; McEwen et al. 2012). Additionally, numer-
ous studies have revealed the significant effects of estrogens on cognition (Fortress
and Frick 2014; Vahaba and Remage-Healey 2015; Sheppard et al. 2018). It has also
been found that estrogen shows protective effects on neurons to counteract the
toxicity of amyloid plaques in Alzheimer’s disease patients (Thomas et al. 1999).
However, this finding is contradicted by the results from the Women’s Health
Initiative Memory Study, which reported that combination therapy of estrogen
with progestin increased the possiblity of acquiring dementia in postmenopausal
women (Shumaker et al. 2003; Webber et al. 2005).
Estrogens also exhibit modulation action on dopaminergic signaling via their D1
receptors potentiation activity and D2 (Dopamine 2) receptor antagonizing activity
(Soares et al. 2003; Hedges et al. 2010). Through their antagonistic effects on D2
receptors, estrogens can lessen the severe symptoms of psychotic disorders. On the
other hand, they may intensify addictive behaviors through their agonistic actions on
D1 receptors (Hedges et al. 2010). Estrogens have modulating activity on other
signaling systems, which includes their stimulating effect on glutamatergic activity
(Cyr et al. 2001), and their negative effect on GABAergic activity (Wójtowicz et al.
2008; Wójtowicz and Mozrzymas 2010). These modulating effects of estrogens on
D1 and D2 receptors are believed to trigger their positive stimulation in mood
management, cognition improvement (Amin et al. 2005) and attention (Soares
et al. 2003; Soares and Frey 2010). Other neuroactive steroids belonging to this
class such as estradiol also display their enhancing effects on the development and
care of the central nervous system (McEwen and Alves 1999). This is clearly evident
in the presence of estrogen receptors in the wide array of brain structures (McEwen
and Alves 1999). Studies have shown that estrogen receptors expressed on
GABAergic neurons may play a key role by which estradiol exerts an excitatory
effect on the nervous system (Schultz et al. 2009). Additionally, it is demonstrated
that positive modulation of ERα on GABAergic neurons decreases the release of
GABA, the inhibitory neurotransmitter (Xiao et al. 2003).
known to play a vital role in the process of synapse formation in the spine (Leranth
et al. 2004; Romeo et al. 2005).
Several studies have demonstrated testosterone’s ability to induce positive mod-
ulatory effects on serotonergic signaling indirectly when it is metabolized to estra-
diol, thereby activating the estrogen receptors (Ebinger et al. 2009). Testosterone
also synergistically enhances the effect of noradrenergic antidepressant agents
through its androgen receptor activating activity (Martı́nez-Mota and Fernández-
Guasti 2004; Ebinger et al. 2009). Similarly, DHEA and its sulfated derivative have
an affinity with a variety of receptors including GABAA, sigma-1 and metabotropic
glutamate receptor (mGluR) (Maninger et al. 2009). Owing to its versatility of action
toward various receptors, DHEA and its sulfated derivative can induce various
synaptic transmissions including GABAergic, dopaminergic, cholinergic and
glutamatergic synaptic transmission (Yoon et al. 2010; Xu et al. 2012). In the
nervous tissues, DHEA is believed to affect neuronal excitability via its modulating
effect on the NMDA receptor (Baulieu 1997) and its positive allosteric modulating
effect on the GABAA receptor (Marx et al. 2006). This is confirmed through an
animal model study which demonstrated that DHEA and its sulfated derivative can
bring about excitation of neuronal function in the nervous system of rodents mainly
by their ability to activate the glutamate and GABA-releasing neurons (Wolf and
Kirschbaum 1999; Meyer et al. 2002; Dong et al. 2007). DHEA also displays
neuroprotective and anti-glucocorticoid effects, thereby aiding in the improvement
of depression, anxiousness, psychotic symptoms and cognitive deficits (Maninger
et al. 2009). DHEA and its sulfated derivative have a protective effect against
NMDA-induced neurotoxicity. The pathophysiological activity of DHEA and its
sulfated derivative also include neuroprotection, enhancing neuronal survival,
neurogenesis and neurite growth (Pluchino et al. 2015). Despite its modulating
effect on various synaptic transmissions and their neurotransmitter production,
DHEA sulfate can also activate the postsynaptic receptors, thereby, encouraging
various significant brain activities, including antidepressant and anxiolytic effects
and enhancement of memory, especially in patients suffering from brain function
disorders such as Alzheimer’s disease and addiction (Kaminska et al. 2000;
Balashov 2010; Dong and Zheng 2012).
increased risk for developing mental health disorders including anxiety, post-
traumatic stress disorder and depression. Numerous studies has revealed that corti-
costerone pretreatment before stress, as observed in rodent models, helps in proper
functioning of the HPA axis, which would reduce stress and stimulate improved
emotional stability (Whitaker et al. 2016). Glucocorticoids such as cortisol have
been demonstrated to enhance regulatory activity in the ventro-lateral prefrontal
cortex. Research also suggests that delayed cortisol release in response to a stressor
facilitates cognitive emotion regulation processes that might be beneficial for restor-
ing emotional stability in the aftermath of stressful events (Jentsch et al. 2019). In
recent studies it has been found that cortisol concentration level is high in patients
suffering from psychotic disorders including schizophrenia. Cortisol concentration
level has also been found to be closely linked to disorders such as cognitive
dysfunction, mild cognitive impairment and Alzheimer’s disease (Mondelli et al.
2015; Pietrzak et al. 2017; Hubbard and Miller 2019). The positive results of various
investigations advocate the possibility of therapies that target depressing cortisol and
Aβ levels, which can be employed in extenuating cognitive impairment in
Alzheimer’s disease. Additionally, in the case of the commencement of psychosis,
cortisol can act as a predictor of treatment response or can be considered as a target
for the design of new therapeutic agents (Mondelli et al. 2015; Hubbard and Miller
2019).
The physiological role of other neuroactive steroids, for example pregnenolone
and pregnenolone sulfate, also has implications for their role in neuronal excitability
and plasticity. Pregnenolone, the precursor to neuroactive steroid progesterone,
possesses the ability to boost the neurotransmitter activity of GABA and to directly
activate the GABAA receptor, as observed in studies conducted using bovine
preparations (Callachan et al. 1987). Pregnenolone action on GABA receptor can
either bring about prolonged temporal influx of chloride ions or can directly open the
ion channel when there is elevated plasma concentration of pregnenolone. On the
other hand, pregnenolone sulfate has an inhibitory action toward the GABA receptor
and acts as an antagonist, as evidenced in an investigation utilising rodent cell
cultures (Mienville and Vicini 1989). The mechanism of pregnenolone-sulfate
antagonistic activity is due to its ability to cause a decline in the frequency of the
chloride channels opening on the GABA receptor, thereby reducing inhibition of
neuron discharge, ultimately producing an excitatory effect on the nervous system
(Mienville and Vicini 1989).
The neuroactive steroid pregnenolone has also been described to modulate
molecular targets such as sigma1 receptors and the type-1 cannabinoid (CB1)
receptor (Vallée 2016). It has a positive modulating effect on the sigma1 receptors
but exhibits a positive modulative effect on type-1 cannabinoid receptor.
Pregnenolone’s positive modulating activity toward brain activity advocates the
possibility of pregnenolone involvement in processes that are activated through the
CB1 receptor, including anxiety, memory, cognition and reward-related behavior
(Fujiwara and Egashira 2004; Gardner 2005). Some of the neuroactive steroids
which have been marketed or are under investigation for their action against various
neuronal disorders are listed in Table 14.1.
14 Hormones and Steroids as Neurotransmitters 469
Table 14.1 List of marketed and investigational neuroactive steroid drugs for the treatment of
neurological disorders
Sl.
no Drug Indication Brand name
1 Pregnenolone Used for the treatment of Alzheimer’s Pregnenolone, Life-
disease, depression, seizures Flo Pregnenolone,
etc.
2 Allopregnenolone Used for the treatment of postpartum Zulresso
depression
3 DHEA Used for the treatment of Alzheimer’s Intrarosa
disease, schizophrenia
4 Ganaloxone Under development for the treatment of NCT02358538,
(Investigational postpartum depression, uncontrolled NCT03865732
drug) seizures in female children and other rare
genetic epilepsies
5 Zuranolone (SAGE- Under development for the treatment of NCT04007367,
217) major depressive disorder, postpartum NCT02978781,
(Investigational depression, essential tremor, Parkinson’s NCT03000569
drug) disease, insomnia and seizures
6 Mifepristone Under development for the treatment of NCT00867360,
(Investigational psychotic major depression and other NCT00637494,
drug) depressive disorders NCT00146523
O
H HO O
CH3 OH N OH
H CF3 O
N S
H
CH3 CH3 CH3 OH F
OH H
F
H O N H H
H H OH
N H
HO H3C O H H O H H H H
Br S
(1) OH O O CH3 (5) HO O HO
(2)
(3) (4) (6) (7)
O
O O O O O O
O O OH
H
H N
O O H H H H N H H N
H H N N H H
S H H
HO O H H H H H H
O H H CN H H
HO H H
O HO H HO O
(8) H HO
(9)
H
(15)
(11) (12) (13)
(10) (14)
OH CF3
O CH(CH3)2
O O OH CH2 CH2CH2CH2CH3 Cl
O H
H H
H H
H H H H N O O
HO HO
HO HO HO HO HO HO
HO HO OH OH
(16) OH OH
(17) (18) (19)
(21) (22)
(20)
(23)
F F
F
F
O
NH F
F
H H
O N
H H
(24)
Fig. 14.8 Examples of agonists to the neuroactive hormone and steroid receptors and their biological activities
S. Marwein et al.
14 Hormones and Steroids as Neurotransmitters 471
14th day. When dopamine markers in the mice were assessed the results showed that
17β-estradiol inclusion in the therapy prevents the depletion of striatal dopamine and
that MTTP could not cause reduction in the dopamine level in mice (Callier et al.
2001; Grandbois et al. 2000). Likewise, studies have reported that unilateral admin-
istration of 6-hydroxydopamine (6-OHDA) in an ovariectomized female rat model
with Parkinson’s disease can induce similar biochemical alteration and abnormal
behavioural patterns to that seen in human Parkinson’s disease (Baraka et al. 2011).
The administration of 17β-estradiol has been reported to prevent these biochemical
and behavioral alterations induced by 6-OHDA in ovariectomized rats, as observed
in an MPTP-induced mouse model (Quesada et al. 2008; Baraka et al. 2011).
Estradiol when given as an adjuvant has been reported to alleviate psychotic-like
behavioural disorders via its neuroprotective ability (Akhondzadeh et al. 2003;
Begemann et al. 2012; Kulkarni et al. 2008a, b, 2011, 2015).
Likewise, the neuroprotective effects of other estrogen receptor modulators such
as the ERα agonist, propyl-pyrazoletriol (PPT) (2) and raloxifene (3), which acts as a
selective estrogen receptor modulator (SERM) on MPTP-treated mice, are also
described (Grandbois et al. 2000; Callier et al. 2001; Bourque et al. 2019). These
two compounds prevented MPTP-induced striatal dopamine depletion in mice when
the drug treatment was continued for 5–14 days. Similarly, a study on a 6-OHDA
lesion-induced rat model of Parkinson’s disease has shown that raloxifene and
compound 2 prevented the induction of biochemical alterations and altered
behavioural function by 6-OHDA (Baraka et al. 2011).
The examination of raloxifene (3) as a selective modulator of the estrogen
receptor and its ability to prevent the estrogenic stimulation of the gonadal tissue
caused by the long-term use of estradiol has been reported (Chlebowski et al. 2009).
Raloxifene’s ability to selectively modulate the estrogen receptors has also been
reported to enhance memory (Yaffe et al. 2005). In a clinical trial using raloxifene as
an adjuvant therapy for schizophrenia (Kulkarni et al. 2010) it was described that
raloxifene stimulates enhancement of the positive symptoms and improvement of the
negative symptoms in postmenopausal women within a dose of 120 mg/day. Con-
sequent investigations with a dose of 60 mg/day of raloxifene as an adjunct
displayed significant enhancement of cognitive ability and improvement in negative
symptoms (Usall et al. 2011).
The synthetic steroid tibolone (4) is considered as a selective tissue estrogenic
activity regulator due to its ability to convert into neuroactive metabolites that could
bind directly to steroid receptors and regulate enzymes needed for its metabolization
into other neuroactive metabolites depending on the tissue involved (Kloosterboer
2004; Reed and Kloosterboer 2004; Cummings et al. 2008). There are reports which
describe the neuroprotective activity of tibolone in neurons (Belenichev et al. 2012;
Pinto-Almazán et al. 2012; Farfán-García et al. 2014). Tibolone has also been
reported to display protective activity in a human astrocyte cell model whereby it
is first metabolized into 3α-hydroxy tibolone and 3β-hydroxy tibolone followed by
the binding of these metabolites to the ERα and ERβ receptors in a human astrocytes.
It thus results in the activation of these receptors to further bring about the desired
neuronal protective activity (Guzman et al. 2007; Avila Rodriguez et al. 2014).
472 S. Marwein et al.
The neuroactive steroid progesterone (9) has also been reported for its
neuroprotective effects and its ability to enhance neuronal survival in males and
females (Wei and Xiao 2013). This activity of progesterone was reported by
Schumacher et al. (2004) whereby they described the neuroprotection action of
progesterone for the dopaminergic neurons in rats. In this study progesterone
displayed protectective activity for the dopaminergic neurons against MPTP-
induced degeneration, an outcome which is closely related to hormonal-induced
weakening of dopaminergic signaling caused by age and the onset of Parkinson’s
disease (Schumacher et al. 2004). Additionally, the beneficial effects of progesterone
for the treatment of multiple sclerosis is evidenced in the experimental autoimmune
encephalomyelitis model. Progesterone exerts its action through its involvement in
repairing of the myelin sheath and by increasing the concentration of endogenous
oligodendrocyte precursor cells (Zawadzka and Franklin 2007).
Progestin (10), a synthetic form of progesterone, is also known for its association
with physiological processes including preservation of myelin sheath integrity,
formation of synapses and neuronal survival (McEwen and Woolley 1994; Mellon
2007; Zhang et al. 2010; Schumacher et al. 2012). Progestin has been reported to
display neuroprotection action in the case of spinal cord and peripheral nerve injury
(Labombarda and Garcia-Ovejero 2014). There are also data which support the
neuroprotective effect of progestin in brain disorders that include demyelinating
disease, motor neuron diseases, epilepsy and depression (Frye and Lacey 2000; Walf
et al., 2006; Deutsch et al. 2013; Li et al. 2013).
Allopregnanolone (11), a metabolic product of progesterone, is a well-known
endogenous neurosteroid with inhibitory action. This neuroactive steroid acts as an
allosteric modulator toward the GABAA receptor and exerts its modulative
properties at the synaptic and extrasynaptic GABAA receptors. Allopregnanolone
is well known for its anticonvulsion property, and numerous experimental animal
models of epilepsy such as the pentylenetetrazol (PTZ)-induced seizure model and
picrotoxin-induced and bicuculline-induced seizure models have helped to establish
this fact (Kaminski et al. 2004; Reddy and Rogawski 2012; Rogawski et al. 2013).
The anticonvulsant activity of allopregnanolone is observed to fully suppress limbic
seizures and status epilepticus in animal models induced by pilocarpine and kainic
acid (Reddy 2010, 2011; Reddy and Rogawski 2012). A phase III clinical trial on the
anticonvulsant activity of allopregnanolone revealed that continuous parenteral
infusion of allopregnanolone formulation represents a better therapy regime for the
management of persistent seizure, a deadly brain condition known as super refrac-
tory status epilepticus (SRSE), that does not respond to usual treatments
(Vaitkevicius et al. 2013; Bialer et al. 2015). Allopregnanolone has also been
reported to have better effects in the management of postpartum depression (PPD)
and essential tremor (ET) in a phase II clinical trial and exploratory study, respec-
tively (Ellenbogen et al. 2016; Kanes et al. 2017a). In the case of postpartum
depression it is observed that with the delivery of the placenta, allopregnanolone
levels decrease rapidly, and this is believed to cause the onset of postpartum
depression in susceptible women (Smith et al. 2007). This is not the case in a healthy
women, as the rapid decline in allopregnanolone levels activates GABA receptor
474 S. Marwein et al.
seizures in adult patients (NCT01963208 n.d.). This study established safety and
tolerability issues with the revelation of some adverse events such as drowsiness and
fatigue. Another ongoing clinical study on ganaxolone is the phase II multicenter,
open-label, proof-of-concept study for the management of epilepsy in children
(NCT02358538 n.d.).
The neuroactive steroid testosterone (15) acting as an agonist to the androgen
receptors was also reported to have a neuroprotective effect (Moffat et al. 2002). The
Baltimore Longitudinal Study of Aging (BLSA) reported significant results regard-
ing the neuroprotection action of testosterone in the case of deterioration of cognitive
ability in Alzheimer’s disease due to aging. The study reports that elevated endoge-
nous testosterone concentration can stimulate verbal memory enhancement in young
patients which is otherwise absent in elders and in men with testosterone deficiency
(Moffat et al. 2002). Studies also report that low testosterone production in the case
of older men increases the risk for the onset of Parkinson-like disorders (Okun et al.
2006). Accordingly, many investigations have been conducted which have described
the benefit of including testosterone in therapy meant to correct Parkinson disease-
like symptoms due to its ability to directly effect and improve motor symptoms
(Okun et al. 2002). Mitchell and group noted the substantial decrease of resting
tremor and improved motor symptoms in a Parkinson’s patient with testosterone
deficiency only after the administration of testosterone (Mitchell et al. 2006).
The neuroactive steroid DHEA (16) and its role in alleviating depressive
symptoms have been studied widely. Recent studies have revealed that higher
DHEA and DHEA sulfate levels prevent the commencement of depressive
symptoms in both males and females (Veronese et al. 2015). However,
low-plasma level DHEA sulfate was described to alleviate severe depression in
men but not in women (Veronese et al. 2015). Additionally, a longitudinal cohort
study revealed that low levels of DHEA sulfate improve depressive symptoms
irrespective of gender and age (Souza-Teodoro et al. 2016).
DHEA analogs conjugated with spiro-epoxy functional group at C-17 were found
to induce substantial protective activity on neurons. Compounds under this class act
as good positive modulators of ion channel receptors such as the NMDA and
GABAA receptors. The spiro-epoxy steroidal derivatives 17–19 reported by
Calogeropoulou et al. (2009) were found to be the most potent neuroprotective
agents against induced apoptosis when tested in neural PC12 cells. In addition,
treatment of PC12 cells with neuroactive steroid derivatives 17–19 also up-regulates
the production of dopamine from the dopaminergic neurons. A further structure
activity relationship study showed that introduction of an epoxide moiety at C-17, as
in the case of the DHEA analogs 17–19, facilitates their easy transportation across
the cell membrane, whereby they can bind directly to estrogen receptors ERɑ and
ERβ. The structure activity relationship study also revealed that the presence of the
hydroxy group at C-21 in 18 and 19 encourages better hydrogen bond formation and
better interaction with the NMDA or the GABAA receptors that can be credited with
enhancing neuroprotective activity of these spiro-epoxy conjugated steroids
(Calogeropoulou et al. 2009).
476 S. Marwein et al.
Alternative hydroxy derivatives of DHEA (20–22) that are known to bind to the
membrane receptors, NMDA and GABAA receptors with high affinity, were studied
for their neuroprotective effects against hypoxia-induced neurodegeneration in the
cortical neurons. The triol derivatives of DHEA could easily cross any physiological
membrane, then directly bind to the membrane receptors and display significant
neuroprotection of the neurons. Supplementary structure activity relationship studies
revealed that addition of a methylene functional group at C-17 as in the derivative 20
produces less neuroprotective effect, while incorporation of the alkyl moiety at C-17
just as in the derivatives 21 and 22 presented improved dimensional flexibility and
enhanced interaction with NMDA and GABAA receptors, leading to better
neuroprotective effects (Long et al. 2016).
The compound 1OP-2198 or XEN 1101 (undisclosed structure) represents a new
class of neuronal allosteric modulator of the Kv7 (KCNQ) potassium channel. The
Kv7 neuronal channels have been described for their function of slowly activating
and deactivating voltage-gated M-current. Studies validate the role these channels
play in the management of hyperexcitability disorders such as epilepsy (Large et al.
2012). 1OP-2198 has been described to exhibit anticonvulsant effect in the maximal
electroshock seizure, in pentylenetetrazol-induced, picrotoxin-induced, bicuculline-
induced, and 6 Hz seizure animal models (Hoffmann et al. 2017). Studies reported
that the 1OP-2198 mechanism of action is comparable to that of other Kv7 channel
modulators such as retigabine; however, it is more potent than the other modulators
(Roeloffs et al. 2008). 1OP-2198 lessens the state of convulsion, thereby exerting
antiepileptic action through a mechanism which involves opening of the neuronal
Kv7 channels through their direct interaction with the ion channel (Roeloffs et al.
2008).
Neuroactive steroids also exert a protective activity on the diabetic-induced
degeneration of neurons. But these steroidal compounds can trigger endocrine side
effects when administered into the physiological system (Moran et al. 2013; Mehlig
et al. 2014). However, other physiological pathways could be targeted so as to
induce the neuroactive steroid production directly in the nervous tissues specifically;
for example, the liver X receptors (LXRs) could be targeted instead of the conven-
tional steroid receptors. In this regard, the LXR agonist GW3965 (23) was evaluated
and reported to increase the neuroactive steroid level specifically in nervous tissues
such as the cerebral cortex, cerebellum and spinal cord of the diabetic-induced rat
model (Mitro et al. 2012). Studies also demonstrated that the LXR agonist GW3965
stimulates an upsurge production of progesterone metabolites that include
dihydroprogesterone, tetrahydroprogesterone and isopregnanolone, and these
metabolites themselves are potent GABAA receptor modulators. In particular,
isopregnanolone does not interact or modulate the GABAA receptor activity, but it
exhibits an antagonistic effect against the tetrahydroprogesterone-activation of the
GABAA receptor (Bäckström et al. 2005). These findings suggest that LXR agonist
GW3965 can improve neuronal synaptic activity in diabetic patients through its
indirect activation of GABAA receptors (Mitro et al. 2012).
Some of the neuroactive compounds exert their neuronal activity not by their
interaction with any receptors but by targeting the enzymes that are necessary for the
14 Hormones and Steroids as Neurotransmitters 477
biosynthesis of steroids rather than acting directly on the receptors involved in the
neurotransmission functions of the neurotransmitters. For instance, the 5α-reductase
inhibitor dutasteride (24) has been described for its ability to activate dopaminergic
signaling and as a drug beneficial in the management of motor neuron disorders and
other neurodegenerative disorders including Parkinson’s disease (Paba et al. 2011).
Current research shows that dutasteride prevented the MPTP-induced damage of
several dopaminergic markers in male mice (Litim et al. 2015). The study was
conducted on mice which were previously exposed to MPTP so as to reduce the
striatal dopamine in the mice brain up to 50%. However, the MPTP-exposed mice
that received dutasteride showed a significant increase in striatal dopamine and its
metabolite concentrations. Dutasteride prevented MPTP toxicity on dopamine
metabolism in MPTP-treated mice (Litim et al. 2015). The neuroprotective activity
of dutasteride could be facilitated by its 5ɑ-reductase inhibitory action, which in turn
stops the conversion of these steroid precursors into their metabolites, leading to
buildup of the precursors such as progesterone, estrogens and DHEA which are
known to possess a neuroprotective effect against MPTP-induce neurodegeneration
(Bourque et al. 2009; Bourque et al. 2019).
The antagonists to the hormone and steroid receptors include drugs and derivatives
of various structural functionality, acting on the different receptors, namely, GABAA
receptors (Johansson et al. 2015), NMDA receptors (Hu et al. 2014), progestin
receptors (Sun et al. 2018), AMPA glutamate receptors (French et al. 2012) and
various ion channels including the voltage-gated sodium channels (Bialer et al.
2013), T-type calcium channels (Tringham et al. 2012) and Na(+)-K(+)-2Cl()
cotransporter (Löscher, Puskarjov & Kaila 2013) that are involved in the neurotrans-
mitter signaling of the hormone and steroidal neurotransmitter. Through their antag-
onistic effects, they exert various therapeutic effects on various neurological
disorders. Examples of antagonists to the neuroactive hormone and steroid receptors
are shown in Fig. 14.9.
There are a group of neuroactive steroid compounds that exhibit an antagonistic
effect toward the GABAA receptor and these are categorized as the GABAA receptor
modulating steroid antagonists (GAMSA). They selectively inhibit neuroactive
steroid-mediated enhancement of GABA-evoked currents at the GABAA receptor.
These compounds exert an antagonistic action to the activation effect of the GABAA
receptor by the neurosteroid but do not hamper the neurotransmitter GABA to
induce chloride flux via the receptor (Johansson et al. 2016). The most common
example of GAMSA is the neuroactive steroid isoallopregnanolone (25), similar to
allopregnanolone, whereby the difference is in the hydroxyl group orientation at C-3
in the A-ring of the steroid. Its antagonistic action toward the GABAA receptors has
been extensively explored through various experimental models. For instance, the
voltage-clamp model using the recombinant GABAA receptors containing rat
478 S. Marwein et al.
H3C
O OH N OH
H H
H
H H H H
H H
HO HO HO
HO H H
H
(25) (26) (27) (28)
H H
O O
O H H
O
H F H H
H2N
O
H H H
HO
HO
OH
(29) COOH
(30) (31)
CH2
O CH3 H3C
N
HO H3C
OH
O O
HO C (CH2)n C O H
H HO
HO
n=1-6 OH H
O
(32)
(33) (34)
Fig. 14.9 Examples of antagonists to the neuroactive hormone and steroid receptors and their
biological activities
O OH H2N
O SiMe3
N
N S
N
HO
HO HO
OH
NO2 CN
O O
MeO MeO
MeO H H
MeO H
H
H H H H H
H H
H H HO HO
O H H
O H
(38) (39) (40) (41)
Fig. 14.10 Examples of patented neuroactive steroids and their biological activities
and are considered valuable in the treatment and/or prevention of various CNS
disorders including epilepsy, cognitive dysfunction, depressive or bipolar disorders,
stroke and multiple sclerosis (Hogenkamp 2014). Similarly, Covey and Robichaud
patented their invention of the neuroactive 19-alkoxy-17-substituted steroids 38–41
for their effectiveness against CNS disorders through their action on the GABA
receptors regulating the excitation of neuron, mood disorder and decreasing stress
level (Covey and Robichaud 2014). Examples of patented neuroactive steroids are
shown in Fig. 14.10.
Neuroactive steroids embodied various protective therapeutic agents against CNS
(central nervous system) and PNS (peripheral nervous system) disorders and
diseases. Nevertheless, attention must be given to other aspects that are associated
with the application of neuroactive steroids. It is important to point out that system-
atic administration of neuroactive steroids can bring about endocrine side effects.
Undeniably, the receptors for neuronal excitability, including the steroid receptors
themselves, are extensively distributed in several nervous tissues. The study describ-
ing the SERMs provides an example of alternative approaches to selectively activate
only those receptors expressed in the nervous tissues. SERMs are molecules that
exert regulation of estrogen receptor and transcriptional activity in specific tissues,
especially in nerve cells, and may not exert any effects or even have an antagonistic
effects toward the estrogen receptors expressed in other cell types (Arévalo et al.
2015). However, the use of synthetic steroids may not fully evoke the desired effects
as the actions of neuroactive steroids depend on their conversion into active
metabolites. Most of the synthetic steroidal receptor ligands cannot be metabolized
in other active metabolites and are ineffective or induce partial effects. This occurs
with medroxyprogesterone acetate whereby it is not converted to the progesterone
metabolites (i.e., dihydroprogesterone and tetrahydroprogesterone) in the CNS as is
natural progesterone (Ciriza et al. 2006).
482 S. Marwein et al.
14.8 Conclusions
References
Akhondzadeh S, Nejatisafa AA, Amini H, Mohammadi MR, Larijani B, Kashani L, Raisi F,
Kamalipour A (2003) Adjunctive estrogen treatment in women with chronic schizophrenia: a
double-blind, randomized, and placebo-controlled trial. Progr Neuropsychopharmacol Biol
Psychiatry 27(6):1007–1012
Akk G, Shu HJ, Wang C, Steinbach JH, Zorumski CF, Covey DF, Mennerick S (2005)
Neurosteroid access to the GABAA receptor. J Neurosci 25(50):11605–11613
Akk G, Covey DF, Evers AS, Steinbach JH, Zorumski CF, Mennerick S (2007) Mechanisms of
neurosteroid interactions with GABAA receptors. Pharmacol Ther 116(1):35–57
Akk G, Covey DF, Evers AS, Steinbach JH, Zorumski CF, Mennerick S (2009) The influence of the
membrane on neurosteroid actions at GABAA receptors. Psychoneuroendocrinology 34:S59–
S66
Almey A, Filardo EJ, Milner TA, Brake WG (2012) Estrogen receptors are found in glia and at
extranuclear neuronal sites in the dorsal striatum of female rats: evidence for cholinergic but not
dopaminergic colocalization. Endocrinology 153(11):5373–5383
14 Hormones and Steroids as Neurotransmitters 483
Bianchi MT, Macdonald RL (2002) Slow phases of GABAA receptor desensitization: structural
determinants and possible relevance for synaptic function. J Physiol 544(1):3–18
Bianchi MT, Macdonald RL (2003) Neurosteroids shift partial agonist activation of GABAA
receptor channels from low-to high-efficacy gating patterns. J Neurosci 23(34):10934–10943
Birzniece V, Bäckström T, Johansson IM, Lindblad C, Lundgren P, Löfgren M, Olsson T,
Ragagnin G, Taube M, Turkmen S, Wahlström G (2006) Neuroactive steroid effects on
cognitive functions with a focus on the serotonin and GABA systems. Brain Res Rev 51
(2):212–239
Bixo M (2014) Biomarkers for premenstrual dysphoric disorder. GABAA modulating steroid
antagonists—a possible treatment for premenstrual dysphoric disorder. In 44th Annual Meeting
of the International Society of Psychoneuroendocrinology, 19–22 August
Bologa CG, Revankar CM, Young SM, Edwards BS, Arterburn JB, Kiselyov AS, Parker MA,
Tkachenko SE, Savchuck NP, Sklar LA, Oprea TI (2006) Virtual and biomolecular screening
converge on a selective agonist for GPR30. Nat Chem Biol 2(4):207
Boonyaratanakornkit V (2011) Scaffolding proteins mediating membrane-initiated extra-nuclear
actions of estrogen receptor. Steroids 76(9):877–884
Boonyaratanakornkit V, McGowan E, Sherman L, Mancini MA, Cheskis BJ, Edwards DP (2007)
The role of extranuclear signaling actions of progesterone receptor in mediating progesterone
regulation of gene expression and the cell cycle. Mol Endocrinol 21(2):359–375
Boulware MI, Mermelstein PG (2009) Membrane estrogen receptors activate metabotropic gluta-
mate receptors to influence nervous system physiology. Steroids 74(7):608–613
Boulware MI, Weick JP, Becklund BR, Kuo SP, Groth RD, Mermelstein PG (2005) Estradiol
activates group I and II metabotropic glutamate receptor signaling, leading to opposing
influences on cAMP response element-binding protein. J Neurosci 25(20):5066–5078
Boulware MI, Heisler JD, Frick KM (2013) The memory-enhancing effects of hippocampal
estrogen receptor activation involve metabotropic glutamate receptor signaling. J Neurosci 33
(38):15184–15194
Brinton RD, Thompson RF, Foy MR, Baudry M, Wang J, Finch CE, Morgan TE, Pike CJ, Mack
WJ, Stanczyk FZ, Nilsen J (2008) Progesterone receptors: form and function in brain. Front
Neuroendocrinol 29(2):313–339
Brown N, Kerby J, Bonnert TP, Whiting PJ, Wafford KA (2002) Pharmacological characterization
of a novel cell line expressing human α4β3δ GABAA receptors. Br J Pharmacol 136
(7):965–974
Brown ES, Park J, Marx CE, Hynan LS, Gardner C, Davila D, Nakamura A, Sunderajan P, Lo A,
Holmes T (2014) A randomized, double-blind, placebo-controlled trial of pregnenolone for
bipolar depression. Neuropsychopharmacology 39(12):2867
Bu J, Zu H (2014) Effects of pregnenolone intervention on the cholinergic system and synaptic
protein 1 in aged rats. Int J Neurosci 124(2):117–124
Callier S, Morissette M, Grandbois M, Pélaprat D, Di Paolo T (2001) Neuroprotective properties of
17β-estradiol, progesterone, and raloxifene in MPTP C57Bl/6 mice. Synapse 41(2):131–138
Cai H, Zhou X, Dougherty GG, Reddy RD, Haas GL, Montrose DM, Keshavan M, Yao JK (2018)
Pregnenolone-progesterone-allopregnanolone pathway as a potential therapeutic target in first-
episode antipsychotic-naïve patients with schizophrenia. Psychoneuroendocrinology 90:43–51
Callachan H, Cottrell GA, Hather NY, Lambert JJ, Nooney JM, Peters JA (1987) Modulation of the
GABAA receptor by progesterone metabolites. Proc R Soc London Ser B Biol Sci 231
(1264):359–369
Calogeropoulou T, Avlonitis N, Minas V, Alexi X, Pantzou A, Charalampopoulos I, Zervou M,
Vergou V, Katsanou ES, Lazaridis I, Alexis MN (2009) Novel dehydroepiandrosterone
derivatives with antiapoptotic, neuroprotective activity. J Med Chem 52(21):6569–6587
Cardona-Gomez GP, DonCarlos L, Garcia-Segura LM (2000) Insulin-like growth factor I receptors
and estrogen receptors colocalize in female rat brain. Neuroscience 99(4):751–760
Carmeci C, Thompson DA, Ring HZ, Francke U, Weigel RJ (1997) Identification of a gene
(GPR30) with homology to the G-protein-coupled receptor superfamily associated with estro-
gen receptor expression in breast cancer. Genomics 45(3):607–617
14 Hormones and Steroids as Neurotransmitters 485
Carré JM, McCormick CM, Hariri AR (2011) The social neuroendocrinology of human aggression.
Psychoneuroendocrinology 36(7):935–944
Carrer HF, Araque A, Buño W (2003) Estradiol regulates the slow Ca2+-activated K+ current in
hippocampal pyramidal neurons. J Neurosci 23(15):6338–6344
Carswell HVO, Dominiczak AF, Macrae IM (2000) Estrogen status affects sensitivity to focal
cerebral ischemia in stroke-prone spontaneously hypertensive rats. Am J Physiol Heart Circ
Physiol 278(1):H290–H294
Carver CM, Reddy DS (2013) Neurosteroid interactions with synaptic and extrasynaptic GABA A
receptors: regulation of subunit plasticity, phasic and tonic inhibition, and neuronal network
excitability. Psychopharmacology (Berl) 230(2):151–188
Cato AC, Nestl A, Mink S (2002) Rapid actions of steroid receptors in cellular signaling pathways.
Sci STKE 2002(138):re9
Chaban VV, Lakhter AJ, Micevych P (2004) A membrane estrogen receptor mediates intracellular
calcium release in astrocytes. Endocrinology 145(8):3788–3795
Charalampopoulos I, Remboutsika E, Margioris AN, Gravanis A (2008) Neurosteroids as
modulators of neurogenesis and neuronal survival. Trends Endocrinol Metab 19(8):300–307
Chen C, Kuo J, Wong A, Micevych P (2014) Estradiol modulates translocator protein (TSPO) and
steroid acute regulatory protein (StAR) via protein kinase A (PKA) signaling in hypothalamic
astrocytes. Endocrinology 155(8):2976–2985
Chen J, Hu R, Ge H, Duanmu W, Li Y, Xue X, Hu S, Feng H (2015) G-protein-coupled receptor
30-mediated antiapoptotic effect of estrogen on spinal motor neurons following injury and its
underlying mechanisms. Mol Med Rep 12(2):1733–1740
Cheng J, Watkins SC, Walker WH (2007) Testosterone activates mitogen-activated protein kinase
via Src kinase and the epidermal growth factor receptor in sertoli cells. Endocrinology 148
(5):2066–2074
Cheng ZX, Lan DM, Wu PY, Zhu YH, Dong Y, Ma L, Zheng P (2008) Neurosteroid dehydroepi-
androsterone sulphate inhibits persistent sodium currents in rat medial prefrontal cortex via
activation of sigma-1 receptors. Exp Neurol 210(1):128–136
Cheskis BJ (2004) Regulation of cell signalling cascades by steroid hormones. J Cell Biochem 93
(1):20–27
Chlebowski RT, Kuller LH, Prentice RL, Stefanick ML, Manson JE, Gass M, Aragaki AK, Ockene
JK, Lane DS, Sarto GE, Rajkovic A (2009) Breast cancer after use of estrogen plus progestin in
postmenopausal women. N Engl J Med 360(6):573–587
Ciriza I, Carrero P, Frye CA, Garcia-Segura LM (2006) Reduced metabolites mediate
neuroprotective effects of progesterone in the adult rat hippocampus. The synthetic progestin
medroxyprogesterone acetate (Provera) is not neuroprotective. J Neurobiol 66(9):916–928
Coleman KM, Smith CL (2001) Intracellular signaling pathways: nongenomic actions of estrogens
and ligand-independent activation of estrogen receptors. Front Biosci 6(1):D1379–D1391
Corcoran C, Walker E, Huot R, Mittal V, Tessner K, Kestler L, Malaspina D (2003) The stress
cascade and schizophrenia: etiology and onset. Schizophr Bull 29(4):671–692
Corpechot C, Robel P, Axelson M, Sjövall J, Baulieu EE (1981) Characterization and measurement
of dehydroepiandrosterone sulfate in rat brain. Proc Natl Acad Sci 78(8):4704–4707
Costa ET, Soto EE, Cardoso RA, Olivera DS, Valenzuela CF (2000) Acute effects of ethanol on
kainate receptors in cultured hippocampal neurons. Alcohol Clin Exp Res 24(2):220–225
Covey D, Robichaud AJ (2014) Washington University in St Louis and Sage Therapeutics Inc,
2014. Neuroactive 19-alkoxy-17-substituted steroids, prodrugs thereof, and methods of treat-
ment using same. U.S. Patent 20140235600
Covey DF, Evers AS, Mennerick S, Zorumski CF, Purdy RH (2001) Recent developments in
structure–activity relationships for steroid modulators of GABAA receptors. Brain Res Rev 37
(1–3):91–97
Cummings SR, Ettinger B, Delmas PD, Kenemans P, Stathopoulos V, Verweij P, Mol-Arts M,
Kloosterboer L, Mosca L, Christiansen C, Bilezikian J (2008) The effects of tibolone in older
postmenopausal women. N Engl J Med 359(7):697–708
486 S. Marwein et al.
Cyr M, Ghribi O, Thibault C, Morissette M, Landry M, Di Paolo T (2001) Ovarian steroids and
selective estrogen receptor modulators activity on rat brain NMDA and AMPA receptors. Brain
Res Rev 37(1–3):153–161
Darbandi-Tonkabon R, Manion BD, Hastings WR, Craigen WJ, Akk G, Bracamontes JR, He Y,
Sheiko TV, Steinbach JH, Mennerick SJ, Covey DF (2004) Neuroactive steroid interactions
with voltage-dependent anion channels: lack of relationship to GABAA receptor modulation
and anesthesia. J Pharmacol Exp Ther 308(2):502–511
Darnaudéry M, Pallarès M, Piazza PV, Le Moal M, Mayo W (2002) The neurosteroid pregnenolone
sulfate infused into the medial septum nucleus increases hippocampal acetylcholine and spatial
memory in rats. Brain Res 951(2):237–242
Dastgheib MONA, Dehpour AR, Heidari M, Moezi L (2015) The effects of intra-dorsal hippocam-
pus infusion of pregnenolone sulfate on memory function and hippocampal BDNF mRNA
expression of biliary cirrhosis-induced memory impairment in rats. Neuroscience 306:1–9
De Nicola AF, Gonzalez SL, Labombarda F, Deniselle MCG, Garay L, Guennoun R, Schumacher
M (2006) Progesterone treatment of spinal cord injury. J Mol Neurosci 28(1):3–15
Deutsch ER, Espinoza TR, Atif F, Woodall E, Kaylor J, Wright DW (2013) Progesterone’s role in
neuroprotection, a review of the evidence. Brain Res 1530:82–105
Dewing P, Boulware MI, Sinchak K, Christensen A, Mermelstein PG, Micevych P (2007) Mem-
brane estrogen receptor-α interactions with metabotropic glutamate receptor 1a modulate female
sexual receptivity in rats. J Neurosci 27(35):9294–9300
Di S, Itoga CA, Fisher MO, Solomonow J, Roltsch EA, Gilpin NW, Tasker JG (2016) Acute stress
suppresses synaptic inhibition and increases anxiety via endocannabinoid release in the
basolateral amygdala. J Neurosci 36(32):8461–8470
Diamanti-Kandarakis E, Dattilo M, Macut D, Duntas L, Gonos ES, Goulis DG, Gantenbein CK,
Kapetanou M, Koukkou E, Lambrinoudaki I, Michalaki M (2017) Mechanisms in endocrinol-
ogy: aging and anti-aging: a combo-endocrinology overview. Eur J Endocrinol 176(6):R283–
R308
Djebaili M, Guo Q, Pettus EH, Hoffman SW, Stein DG (2005) The neurosteroids progesterone and
allopregnanolone reduce cell death, gliosis, and functional deficits after traumatic brain injury in
rats. J Neurotrauma 22(1):106–118
Do Rego JL, Seong JY, Burel D, Leprince J, Luu-The V, Tsutsui K, Tonon MC, Pelletier G, Vaudry
H (2009) Neurosteroid biosynthesis: enzymatic pathways and neuroendocrine regulation by
neurotransmitters and neuropeptides. Front Neuroendocrinol 30(3):259–301
Dong Y, Zheng P (2012) Dehydroepiandrosterone sulphate: action and mechanism in the brain. J
Neuroendocrinol 24(1):215–224
Dong LY, Cheng ZX, Fu YM, Wang ZM, Zhu YH, Sun JL, Dong Y, Zheng P (2007) Neurosteroid
dehydroepiandrosterone sulfate enhances spontaneous glutamate release in rat prelimbic cortex
through activation of dopamine D1 and sigma-1 receptor. Neuropharmacology 52(3):966–974
Dubrovsky BO (2005) Steroids, neuroactive steroids and neurosteroids in psychopathology. Progr
Neuropsychopharmacol Biol Psychiatry 29(2):169–192
Dubrovsky B (2006) Neurosteroids, neuroactive steroids, and symptoms of affective disorders.
Pharmacol Biochem Behav 84(4):644–655
Ebinger M, Sievers C, Ivan D, Schneider HJ, Stalla GK (2009) Is there a neuroendocrinological
rationale for testosterone as a therapeutic option in depression? J Psychopharmacol 23
(7):841–853
Ellenbogen A, Raines S, Kanes S (2016) Exploratory trial results for SAGE-547 in essential tremor
(P4. 297)
Ellsworth KP, Azzolina BA, Cimis G, Bull HG, Harris GS (1998) Cloning, expression and
characterization of rhesus macaque types 1 and 2 5alpha-reductase: evidence for mechanism-
based inhibition by finasteride. J Steroid Biochem Mol Biol 66(5–6):271–279
Evans RM (1988) The steroid and thyroid hormone receptor superfamily. Science 240
(4854):889–895
14 Hormones and Steroids as Neurotransmitters 487
Herrick SP, Waters EM, Drake CT, McEwen BS, Milner TA (2006) Extranuclear estrogen receptor
beta immunoreactivity is on doublecortin-containing cells in the adult and neonatal rat dentate
gyrus. Brain Res 1121(1):46–58
Higo S, Hojo Y, Ishii H, Komatsuzaki Y, Ooishi Y, Murakami G, Mukai H, Yamazaki T,
Nakahara D, Barron A, Kimoto T (2011) Endogenous synthesis of corticosteroids in the
hippocampus. PLoS One 6(7):e21631
Hoffmann E, Wald J, Raines S, Nomikos G, Colquhoun H, Kanes S (2017) The pharmacokinetics
of SAGE-217 in Phase 1 SAD and MAD studies (P3. 008)
Hogenkamp DJ (2014) Novel 17b-heteroaryl-substituted steroids as modulators of gabaa receptors.
U.S. Patent Application 14/166,912
Horishita T, Minami K, Uezono Y, Shiraishi M, Ogata J, Okamoto T, Terada T, Sata T (2005) The
effects of the neurosteroids: pregnenolone, progesterone and dehydroepiandrosterone on mus-
carinic receptor-induced responses in Xenopus oocytes expressing M 1 and M 3 receptors.
Naunyn Schmiedebergs Arch Pharmacol 371(3):221–228
Hu R, Sun H, Zhang Q, Chen J, Wu N, Meng H, Cui G, Hu S, Li F, Lin J, Wan Q (2012) G-protein
coupled estrogen receptor 1 mediated estrogenic neuroprotection against spinal cord injury. Crit
Care Med 40(12):3230–3237
Hu H, Zhou Y, Leng T, Liu A, Wang Y, You X, Chen J, Tang L, Chen W, Qiu P, Yin W (2014) The
major cholesterol metabolite cholestane-3β, 5α, 6β-triol functions as an endogenous
neuroprotectant. J Neurosci 34(34):11426–11438
Hubbard DB, Miller BJ (2019) Meta-analysis of blood cortisol levels in individuals with first-
episode psychosis. Psychoneuroendocrinology. https://doi.org/10.1016/j.psyneuen.2019.03.
014
Huss B, Long NP, Ponce L, Gomez L, Moore CA, Sinchak K (2011) Progesterone rapidly facilitates
sexual receptivity through deactivation of muopioid receptors in the medial preoptic nucleus of
the hypothalamus. In: Society for Neuroscience, Program 391.307, Abstract Viewer/Itinerary
Planner, Washington, DC
Jentsch VL, Merz CJ, Wolf OT (2019) Restoring emotional stability: cortisol effects on the neural
network of cognitive emotion regulation. Behav Brain Res 374:111880
Jiang CS, Guo XJ, Gong JX, Zhu TT, Zhang HY, Guo YW (2012) Synthesis and biological
evaluation of 21-arylidenepregnenolone derivatives as neuroprotective agents. Bioorg Med
Chem Lett 22(6):2226–2229
Johansson IM, Birzniece V, Lindblad C, Olsson T, Bäckström T (2002) Allopregnanolone inhibits
learning in the Morris water maze. Brain Res 934(2):125–131
Johansson M, Agusti A, Llansola M, Montoliu C, Strömberg J, Malinina E, Ragagnin G,
Doverskog M, Bäckström T, Felipo V (2015) GR3027 antagonizes GABAA receptor-
potentiating neurosteroids and restores spatial learning and motor coordination in rats with
chronic hyperammonemia and hepatic encephalopathy. Am J Physiol Gastrointest Liver Physiol
309(5):G400–G409
Johansson M, Strömberg J, Ragagnin G, Doverskog M, Bäckström T (2016) GABAA receptor
modulating steroid antagonists (GAMSA) are functional in vivo. J Steroid Biochem Mol Biol
160:98–105
Joksovic PM, Covey DF, Todorovic SM (2007) Inhibition of T-type calcium current in the reticular
thalamic nucleus by a novel neuroactive steroid. Ann N Y Acad Sci 1122(1):83–94
Jensen EV (1962) On the mechanism of estrogen action. Perspec Biol Med 6(1):47–60
Kelley SP, Alan JK, O’Buckley TK, Mennerick S, Krishnan K, Covey DF, Morrow AL (2007)
Antagonism of neurosteroid modulation of native γ-aminobutyric acid receptors by (3α, 5α)-17-
phenylandrost-16-en-3-ol. Eur J Pharmacol 572(2–3):94–101
Kaminska M, Harris J, Gijsbers K, Dubrovsky B (2000) Dehydroepiandrosterone sulfate (DHEAS)
counteracts decremental effects of corticosterone on dentate gyrus LTP. Implications for
depression. Brain Res Bull 52(3):229–234
490 S. Marwein et al.
Kaminski RM, Livingood MR, Rogawski MA (2004) Allopregnanolone analogs that positively
modulate GABAA receptors protect against partial seizures induced by 6-Hz electrical stimula-
tion in mice. Epilepsia 45(7):864–867
Kanes S, Colquhoun H, Gunduz-Bruce H, Raines S, Arnold R, Schacterle A, Doherty J, Epperson
CN, Deligiannidis KM, Riesenberg R, Hoffmann E (2017a) Brexanolone (SAGE-547 injection)
in post-partum depression: a randomised controlled trial. Lancet 390(10093):480–489
Kanes SJ, Colquhoun H, Doherty J, Raines S, Hoffmann E, Rubinow DR, Meltzer-Brody S (2017b)
Open-label, proof-of-concept study of brexanolone in the treatment of severe postpartum
depression. Human Psychopharmacol 32(2):e2576
Kapras V, Slavickova A, Stastna E, Vyklicky L Jr, Vales K, Chodounska H (2012) Synthesis of
deuterium labeled NMDA receptor inhibitor–20-Oxo-5β-[9, 12, 12-2H3] pregnan-3α-yl-l-
glutamyl 1-ester. Steroids 77(3):282–287
Karki P, Webb A, Zerguine A, Choi J, Son DS, Lee E (2014) Mechanism of raloxifene-induced
upregulation of glutamate transporters in rat primary astrocytes. Glia 62(8):1270–1283
Kelly MJ, Moss RL, Dudley CA (1976) Differential sensitivity of preoptic-septal neurons to
microelectrophoressed estrogen during the estrous cycle. Brain Res 114(1):152–157
Kelly MJ, Rønnekleiv OK, Ibrahim N, Lagrange AH, Wagner EJ (2002) Estrogen modulation of K
+ channel activity in hypothalamic neurons involved in the control of the reproductive axis.
Steroids 67(6):447–456
Kinlein SA, Phillips DJ, Keller CR, Karatsoreos IN (2019) Role of corticosterone in altered
neurobehavioral responses to acute stress in a model of compromised hypothalamic-pituitary-
adrenal axis function. Psychoneuroendocrinology 102:248–255
Kliewer SA, Moore JT, Wade L, Staudinger JL, Watson MA, Jones SA, McKee DD, Oliver BB,
Willson TM, Zetterström RH, Perlmann T (1998) An orphan nuclear receptor activated by
pregnanes defines a novel steroid signaling pathway. Cell 92(1):73–82
Kloosterboer HJ (2004) Tissue-selectivity: the mechanism of action of tibolone. Maturitas
48:30–40
Koenig JI, Kirkpatrick B, Lee P (2002) Glucocorticoid hormones and early brain development in
schizophrenia. Neuropsychopharmacology 27(2):309
Kudwa AE, Michopoulos V, Gatewood JD, Rissman EF (2006) Roles of estrogen receptors α and β
in differentiation of mouse sexual behavior. Neuroscience 138(3):921–928
Kuiper GG, Enmark E, Pelto-Huikko M, Nilsson S, Gustafsson JA (1996) Cloning of a novel
receptor expressed in rat prostate and ovary. Proc Natl Acad Sci 93(12):5925–5930
Kulkarni J, Riedel A, De Castella AR, Fitzgerald PB, Rolfe TJ, Taffe J, Burger H (2001)
Estrogen—a potential treatment for schizophrenia. Schizophr Res 48(1):137–144
Kulkarni J, de Castella A, Fitzgerald PB, Gurvich CT, Bailey M, Bartholomeusz C, Burger H
(2008a) Estrogen in severe mental illness: a potential new treatment approach. Arch Gen
Psychiatry 65(8):955–960
Kulkarni J, Gurvich C, Gilbert H, Mehmedbegovic F, Mu L, Marston N, Gavrilidis E, de Castella A
(2008b) Hormone modulation: a novel therapeutic approach for women with severe mental
illness. Aust N Z J Psychiatry 42(1):83–88
Kulkarni J, Gurvich C, Lee SJ, Gilbert H, Gavrilidis E, de Castella A, Berk M, Dodd S, Fitzgerald
PB, Davis SR (2010) Piloting the effective therapeutic dose of adjunctive selective estrogen
receptor modulator treatment in postmenopausal women with schizophrenia.
Psychoneuroendocrinology 35(8):1142–1147
Kulkarni J, de Castella A, Headey B, Marston N, Sinclair K, Lee S, Gurvich C, Fitzgerald PB,
Burger H (2011) Estrogens and men with schizophrenia: is there a case for adjunctive therapy?
Schizophr Res 125(2–3):278–283
Kulkarni J, Gavrilidis E, Wang W, Worsley R, Fitzgerald PB, Gurvich C, Van Rheenen T, Berk M,
Burger H (2015) Estradiol for treatment-resistant schizophrenia: a large-scale randomized-
controlled trial in women of child-bearing age. Mol Psychiatry 20(6):695–702
Kuo J, Hamid N, Bondar G, Prossnitz ER, Micevych P (2010) Membrane estrogen receptors
stimulate intracellular calcium release and progesterone synthesis in hypothalamic astrocytes.
J Neurosci 30(39):12950–12957
14 Hormones and Steroids as Neurotransmitters 491
Kushida A, Tamura H (2009) Retinoic acids induce neurosteroid biosynthesis in human glial GI-1
Cells via the induction of steroidogenic genes. J Biochem 146(6):917–923
Krȩżel W, Dupont S, Krust A, Chambon P, Chapman PF (2001) Increased anxiety and synaptic
plasticity in estrogen receptor β-deficient mice. Proc Natl Acad Sci 98(21):12278–12282
Löscher W, Puskarjov M, Kaila K (2013) Cation-chloride cotransporters NKCC1 and KCC2 as
potential targets for novel antiepileptic and antiepileptogenic treatments. Neuropharmacol
69:62–74
Labombarda F, Garcia-Ovejero D (2014) Give progesterone a chance. Neural Regen Res 9
(15):1422
Labombarda F, Gonzalez SL, Deniselle MG, Vinson GP, Schumacher M, De Nicola AF, Guennoun
R (2003) Effects of injury and progesterone treatment on progesterone receptor and progester-
one binding protein 25-Dx expression in the rat spinal cord. J Neurochem 87(4):902–913
Labombarda F, Meffre D, Delespierre B, Krivokapic-Blondiaux S, Chastre A, Thomas P, Pang Y,
Lydon JP, Gonzalez SL, De Nicola AF, Schumacher M (2010) Membrane progesterone
receptors localization in the mouse spinal cord. Neuroscience 166(1):94–106
Laconi MR, Reggiani PC, Penissi A, Yunes R, Cabrera RJ (2007) Allopregnanolone modulates
striatal dopamingergic activity of rats under different gonadal hormones conditions. Neurol Res
29(6):622–627
Lambert JJ, Belelli D, Hill-Venning C, Peters JA (1995) Neurosteroids and GABAA receptor
function. Trends Pharmacol Sci 16(9):295–303
Landgren S, Aasly J, Bäckström T, Dubrovsky B, Danielsson E (1987) The effect of progesterone
and its metabolites on the interictal epileptiform discharge in the cat’s cerebral cortex. Acta
Physiol Scand 131(1):33–42
Lapchak PA, Araujo DM (2001) Preclinical development of neurosteroids as neuroprotective
agents for the treatment of neurodegenerative diseases. Int Rev Neurobiol 46:379–397
Large CH, Sokal DM, Nehlig A, Gunthorpe MJ, Sankar R, Crean CS, VanLandingham KE, White
HS (2012) The spectrum of anticonvulsant efficacy of retigabine (ezogabine) in animal models:
implications for clinical use. Epilepsia 53(3):425–436
Le Mellédo JM, Baker GB (2002) Neuroactive steroids, and anxiety disorders. J Psychiatry
Neurosci 27(3):161–165
Lebesgue D, Traub M, De Butte-Smith M, Chen C, Zukin RS, Kelly MJ, Etgen AM (2010) Acute
administration of non-classical estrogen receptor agonists attenuates ischemia-induced hippo-
campal neuron loss in middle-aged female rats. PLoS One 5(1):e8642
Leonhardt SA, Boonyaratanakornkit V, Edwards DP (2003) Progesterone receptor transcription and
non-transcription signaling mechanisms. Steroids 68(10–13):761–770
Leranth C, Hajszan T, MacLusky NJ (2004) Androgens increase spine synapse density in the CA1
hippocampal subfield of ovariectomized female rats. J Neurosci 24(2):495–499
Li Y, Raaby KF, Sanchez C, Gulinello M (2013) Serotonergic receptor mechanisms underlying
antidepressant-like action in the progesterone withdrawal model of hormonally induced depres-
sion in rats. Behav Brain Res 256:520–528
Litim N, Bourque M, Al Sweidi S, Morissette M, Di Paolo T (2015) The 5α-reductase inhibitor
Dutasteride but not Finasteride protects dopamine neurons in the MPTP mouse model of
Parkinson’s disease. Neuropharmacology 97:86–94
Liu SB, Zhang N, Guo YY, Zhao R, Shi TY, Feng SF, Wang SQ, Yang Q, Li XQ, Wu YM, Ma L
(2012) G-protein-coupled receptor 30 mediates rapid neuroprotective effects of estrogen via
depression of NR2B-containing NMDA receptors. J Neurosci 32(14):4887–4900
Long L, Xu L, Xiao Z, Hu S, Luo R, Wang H, Lu X, Xu Z, Yao X, Zhou L, Long H (2016)
Neurological complications and risk factors of cardiopulmonary failure of EV-A71-related
hand, foot and mouth disease. Sci Rep 6:23444
Lösel R, Breiter S, Seyfert M, Wehling M, Falkenstein E (2005) Classic and non-classic progester-
one receptors are both expressed in human spermatozoa. Horm Metab Res 37(01):10–14
492 S. Marwein et al.
Niitsu T, Iyo M, Hashimoto K (2012) Sigma-1 receptor agonists as therapeutic drugs for cognitive
impairment in neuropsychiatric diseases. Curr Pharm Des 18(7):875–883
Oakley RH, Cidlowski JA (2013) The biology of the glucocorticoid receptor: new signaling
mechanisms in health and disease. J Allergy Clin Immunol 132(5):1033–1044
Ogden KK, Traynelis SF (2011) New advances in NMDA receptor pharmacology. Trends
Pharmacol Sci 32(12):726–733
Okun MS, Walter BL, McDonald WM, Tenover JL, Green J, Juncos JL, DeLong MR (2002)
Beneficial effects of testosterone replacement for the nonmotor symptoms of Parkinson disease.
Arch Neurol 59(11):1750–1753
Okun MS, Fernandez HH, Rodriguez RL, Romrell J, Suelter M, Munson S, Louis ED, Mulligan T,
Foster PS, Shenal BV, Armaghani SJ (2006) Testosterone therapy in men with Parkinson
disease: results of the TEST-PD Study. Arch Neurol 63(5):729–735
Orchinik M, Murray TF, Moore FL (1991) A corticosteroid receptor in neuronal membranes.
Science 252(5014):1848–1851
Österlund MK, Witt MR, Gustafsson JÅ (2005) Estrogen action in mood and neurodegenerative
disorders. Endocrine 28(3):235–241
Paradiso K, Sabey K, Evers AS, Zorumski CF, Covey DF, Steinbach JH (2000) Steroid inhibition
of rat neuronal nicotinic α4β2 receptors expressed in HEK 293 cells. Mol Pharmacol 58
(2):341–351
Pluchino N, Luisi M, Lenzi E, Centofanti M, Begliuomini S, Freschi L, Ninni F, Genazzani AR
(2006) Progesterone and progestins: Effects on brain, allopregnanolone and β-endorphin. J
Steroid Biochem Mol Biol 102(1–5):205–213
Paba S, Frau RC, Godar S, Devoto P, Marrosu F, Bortolato M (2011) Steroid 5α-reductase as a
novel therapeutic target for schizophrenia and other neuropsychiatric disorders. Curr Pharm Des
17(2):151–167
Papadopoulos V, Fan J, Zirkin B (2018) Translocator protein (18 kDa): an update on its function in
steroidogenesis. J Neuroendocrinol 30(2):e12500
Paul SM, Purdy RH (1992) Neuroactive steroids. FASEB 6:2311–2322
Pietrzak RH, Laws SM, Lim YY, Bender SJ, Porter T, Doecke J, Ames D, Fowler C, Masters CL,
Milicic L, Rainey-Smith S (2017) Plasma cortisol, brain amyloid-β, and cognitive decline in
preclinical Alzheimer’s disease: a 6-year prospective cohort study. Biol Psychiatry Cogn
Neurosci Neuroimaging 2(1):45–52
Pinto-Almazán R, Calzada-Mendoza CC, Campos-Lara MG, Guerra-Araiza C (2012) Effect of
chronic administration of estradiol, progesterone, and tibolone on the expression and phosphor-
ylation of glycogen synthase kinase-3β and the microtubule-associated protein tau in the
hippocampus and cerebellum of female rat. J Neurosci Res 90(4):878–886
Pisu MG, Serra M (2004) Neurosteroids and neuroactive drugs in mental disorders. Life Sci 74
(26):3181–3197
Pluchino N, Drakopoulos P, Bianchi-Demicheli F, Wenger JM, Petignat P, Genazzani AR (2015)
Neurobiology of DHEA and effects on sexuality, mood and cognition. J Steroid Biochem Mol
Biol 145:273–280
Pratt WB, Toft DO (2003) Regulation of signaling protein function and trafficking by the hsp90/
hsp70-based chaperone machinery. Exp Biol Med 228(2):111–133
Prossnitz ER, Arterburn JB, Smith HO, Oprea TI, Sklar LA, Hathaway HJ (2008) Estrogen
signaling through the transmembrane G protein–coupled receptor GPR30. Annu Rev Physiol
70:165–190
Pappas TC, Gametchu B, Watson CS (1995) Membrane estrogen receptors identified by multiple
antibody labeling and impeded-ligand binding. FASEB J 9(5):404–410
Pathirathna S, Brimelow BC, Jagodic MM, Krishnan K, Jiang X, Zorumski CF, Mennerick S,
Covey DF, Todorovic SM, Jevtovic-Todorovic V (2005) New evidence that both T-type
calcium channels and GABAA channels are responsible for the potent peripheral analgesic
effects of 5α-reduced neuroactive steroids. Pain 114(3):429–443
Pedram A, Razandi M, Sainson RC, Kim JK, Hughes CC, Levin ER (2007) A conserved
mechanism for steroid receptor translocation to the plasma membrane. J Biol Chem 282
(31):22278–22288
496 S. Marwein et al.
Talih F, Fattal O, Malone D (2007) Anabolic steroid abuse: psychiatric and physical costs. Cleve
Clin J Med 74(5):341
Tang YT, Hu T, Arterburn M, Boyle B, Bright JM, Emtage PC, Funk WD (2005) PAQR proteins: a
novel membrane receptor family defined by an ancient7-transmembrane pass motif. J Mol Evol
61(3):372–380
Tang H, Zhang Q, Yang L, Dong Y, Khan M, Yang F, Brann DW, Wang R (2014) GPR30 mediates
estrogen rapid signaling and neuroprotection. Mol Cell Endocrinol 387(1–2):52–58
Thomas T, Rhodin JA, Sutton ET, Bryant MW, Price JM (1999) Estrogen protects peripheral and
cerebral blood vessels from toxicity of Alzheimer peptide amyloid-beta and inflammatory
reaction. J Submicrosc Cytol Pathol 31(4):571–579
Thomas P, Pang Y, Dong J, Groenen P, Kelder JD, De Vlieg J, Zhu Y, Tubbs C (2007) Steroid and
G protein binding characteristics of the seatrout and human progestin membrane receptor α
subtypes and their evolutionary origins. Endocrinology 148(2):705–718
Thorpe L, Whitney DK, Kutcher SP, Kennedy SH (2001) Clinical guidelines for the treatment of
depressive disorders. VI Special populations. Can J Psychiatry 46:63S–76S
Todorovic SM, Pathirathna S, Brimelow BC, Jagodic MM, Ko SH, Jiang X, Nilsson KR, Zorumski
CF, Covey DF, Jevtovic-Todorovic V (2004) 5β-reduced neuroactive steroids are novel voltage-
dependent blockers of T-type Ca2+ channels in rat sensory neurons in vitro and potent
peripheral analgesics in vivo. Mol Pharmacol 66(5):1223–1235
Toft D, Gorski J (1966) A receptor molecule for estrogens: isolation from the rat uterus and
preliminary characterization. Proc Natl Acad Sci U S A 55(6):1574
Torres JM, Ortega E (2003) DHEA, PREG and their sulphate derivatives on plasma and brain after
CRH and ACTH administration. Neurochem Res 28(8):1187–1191
Tringham E, Powell KL, Cain SM, Kuplast K, Mezeyova J, Weerapura M, Eduljee C, Jiang X,
Smith P, Morrison JL, Jones NC (2012) T-type calcium channel blockers that attenuate thalamic
burst firing and suppress absence seizures. Sci Transl Med 4(121):121ra19
Turkmen S, Lundgren P, Birzniece V, Zingmark E, Backstrom T, Johansson IM (2004) 3beta-
20beta-dihydroxy-5alpha-pregnane (UC1011) antagonism of the GABA potentiation and the
learning impairment induced in rats by allopregnanolone. Eur J Neurosci 20:1604–1612
Usall J, Huerta-Ramos E, Iniesta R, Cobo J, Araya S, Roca M, Serrano-Blanco A, Teba F, Ochoa S
(2011) Raloxifene as an adjunctive treatment for postmenopausal women with schizophrenia: a
double-blind, randomized, placebo-controlled trial. J Clin Psychiatry 72(11):1552
van Wingen GA, Ossewaarde L, Bäckström T, Hermans EJ, Fernández G (2011) Gonadal hormone
regulation of the emotion circuitry in humans. Neuroscience 191:38–45
Vahaba DM, Remage-Healey L (2015) Brain estrogen production and the encoding of recent
experience. Curr Opin Behav Sci 6:148–153
Vaitkevicius H, Ng M, Moura L, Rosenthal E, Westover MB, Rosand J, Rogawski MA, Reddy K,
Cole AJ (2013) Successful allopregnanolone treatment of new onset refractory status epilepticus
(NORSE) syndrome: first in man experience. Epilepsia 54(suppl 6):106–124
Valadez-Cosmes P, Vázquez-Martínez ER, Cerbon M, Camacho-Arroyo I (2016) Membrane
progesterone receptors in reproduction and cancer. Mol Cell Endocrinol 434:166–175
Vallée M (2016) Neurosteroids and potential therapeutics: focus on pregnenolone. J Steroid
Biochem Mol Biol 160:78–87
Vallée M, Mayo W, Darnaudéry M, Corpéchot C, Young J, Koehl M, Le Moal M, Baulieu EE,
Robel P, Simon H (1997) Neurosteroids: deficient cognitive performance in aged rats depends
on low pregnenolone sulfate levels in the hippocampus. Proc Natl Acad Sci 94
(26):14865–14870
Vega-Vela NE, Osorio D, Avila-Rodriguez M, Gonzalez J, García-Segura LM, Echeverria V,
Barreto GE (2017) L-type calcium channels modulation by estradiol. Mol Neurobiol 54
(7):4996–5007
Veronese N, De Rui M, Bolzetta F, Zambon S, Corti MC, Baggio G, Toffanello ED, Crepaldi G,
Perissinotto E, Manzato E, Sergi G (2015) Serum dehydroepiandrosterone sulfate and incident
depression in the elderly: the Pro. VA study. Am J Geriatr Psychiatry 23(8):863–871
500 S. Marwein et al.
Walf AA, Frye CA (2006) A review and update of mechanisms of estrogen in the hippocampus and
amygdala for anxiety and depression behavior. Neuropsychopharmacology 31(6):1097
Wang MD, Bäckström T, Landgren S (2000) The inhibitory effects of allopregnanolone and
pregnanolone on the population spike, evoked in the rat hippocampal CA1 stratum pyramidale
in vitro, can be blocked selectively by epiallopregnanolone. Acta Physiol Scand 169
(4):333–341
Wang M, He Y, Eisenman LN, Fields C, Zeng CM, Mathews J, Benz A, Fu T, Zorumski E,
Steinbach JH, Covey DF (2002) 3β-hydroxypregnane steroids are pregnenolone sulfate-like
GABAA receptor antagonists. J Neurosci 22(9):3366–3375
Wang Y, Wang Y, Chen Z (2018) Double-edged GABAergic synaptic transmission in seizures: the
importance of chloride plasticity. Brain Res. https://doi.org/10.1016/j.brainres.2018.09.008
Waters EM, Torres-Reveron A, McEwen BS, Milner TA (2008) Ultrastructural localization of
extranuclear progestin receptors in the rat hippocampal formation. J Comp Neurol 511(1):34–46
Webber KM, Casadesus G, Marlatt MW, Perry G, Hamlin CR, Atwood CS, Bowen RL, Smith MA
(2005) Estrogen bows to a new master: the role of gonadotropins in Alzheimer pathogenesis.
Ann N Y Acad Sci 1052(1):201–209
Wehling M (1997) Specific, nongenomic actions of steroid hormones. Annu Rev Physiol 59
(1):365–393
Wei J, Xiao GM (2013) The neuroprotective effects of progesterone on traumatic brain injury:
current status and future prospects. Acta Pharmacol Sin 34(12):1485
Whitaker AM, Farooq MA, Edwards S, Gilpin NW (2016) Post-traumatic stress avoidance is
attenuated by corticosterone and associated with brain levels of steroid receptor co-activator-1
in rats. Stress 19(1):69–77
Wójtowicz T, Mozrzymas JW (2010) Estradiol and GABAergic transmission in the hippocampus.
Vitam Horm 82:279–300. Academic Press
Wójtowicz T, Lebida K, Mozrzymas JW (2008) 17β-estradiol affects GABAergic transmission in
developing hippocampus. Brain Res 1241:7–17
Wolf OT, Kirschbaum C (1999) Actions of dehydroepiandrosterone and its sulfate in the central
nervous system: effects on cognition and emotion in animals and humans. Brain Res Rev 30
(3):264–288
Xiao L, Jackson LR, Becker JB (2003) The effect of estradiol in the striatum is blocked by ICI
182,780 but not tamoxifen: pharmacological and behavioral evidence. Neuroendocrinology 77
(4):239–245
Xiao L, Qi A, Chen Y (2005) Cultured embryonic hippocampal neurons deficient in glucocorticoid
(GC) receptor: a novel model for studying nongenomic effects of GC in the neural system.
Endocrinology 146(9):4036–4041
Xu Y, Tanaka M, Chen L, Sokabe M (2012) DHEAS induces short-term potentiation via the
activation of a metabotropic glutamate receptor in the rat hippocampus. Hippocampus 22
(4):707–722
Yaffe K, Krueger K, Cummings SR, Blackwell T, Henderson VW, Sarkar S, Ensrud K, Grady D
(2005) Effect of raloxifene on prevention of dementia and cognitive impairment in older
women: the Multiple Outcomes of Raloxifene Evaluation (MORE) randomized trial. Am J
Psychiatry 162(4):683–690
Yagishita T, Kushida A, Tamura H (2012) Vitamin D3 enhances ATRA-mediated neurosteroid
biosynthesis in human glioma GI-1 cells. J Biochem 152(3):285–292
Yan M, Liu AL, Zhou SJ, Tang LP, Ou YQ, Yin W, Chen XY, Su XW, Qiu PX, Huang YJ, Zhang
JX (2015) Characterization of a synthetic steroid 24-keto-cholest-5-en-3β, 19-diol as a
neuroprotectant. CNS Neurosci Ther 21(6):486–495
Yang S, Roselli F, Patchev AV, Yu S, Almeida OF (2013) Non-receptor-tyrosine kinases integrate
fast glucocorticoid signaling in hippocampal neurons. J Biol Chem 288(33):23725–23739
Yoon SY, Roh DH, Seo HS, Kang SY, Moon JY, Song S, Beitz AJ, Lee JH (2010) An increase in
spinal dehydroepiandrosterone sulfate (DHEAS) enhances NMDA-induced pain via
14 Hormones and Steroids as Neurotransmitters 501
phosphorylation of the NR1 subunit in mice: involvement of the sigma-1 receptor. Neurophar-
macology 59(6):460–467
Zakon HH (1998) The effects of steroid hormones on electrical activity of excitable cells. Trends
Neurosci 21(5):202–207
Zawadzka M, Franklin RJ (2007) Myelin regeneration in demyelinating disorders: new
developments in biology and clinical pathology. Curr Opin Neurol 20(3):294–298
Zhang Z, Yang R, Zhou R, Li L, Sokabe M, Chen L (2010) Progesterone promotes the survival of
newborn neurons in the dentate gyrus of adult male mice. Hippocampus 20(3):402–412
Zhao L, O’Neill K, Brinton RD (2006) Estrogenic agonist activity of ICI 182,780 (Faslodex) in
hippocampal neurons: implications for basic science understanding of estrogen signaling and
development of estrogen modulators with a dual therapeutic profile. J Pharmacol Exp Ther 319
(3):1124–1132
Zheng P (2009) Neuroactive steroid regulation of neurotransmitter release in the CNS: action,
mechanism and possible significance. Prog Neurobiol 89(2):134–152
Zhou Q, Sheng M (2013) NMDA receptors in nervous system diseases. Neuropharmacology
74:69–75
Zuloaga DG, Yahn SL, Pang Y, Quihuis AM, Oyola MG, Reyna A, Thomas P, Handa RJ, Mani SK
(2012) Distribution and estrogen regulation of membrane progesterone receptor-β in the female
rat brain. Endocrinology 153(9):4432–4443
Pharmacology of Neuropeptides:
Substance P, Vasoactive Intestinal Peptides, 15
Neuropeptide Y, Calcitonin Peptides
and Their Receptors
Abstract
Keywords
Abbreviations
CD Circular dichroism
CNS Central nervous system
DAG Diacylglycerol
GDP Guanosine diphosphate
GLP Glucagon-like peptide
GPCRs G protein-coupled receptors
GTP Guanosine triphosphate
IL Interleukin
15.1 Introduction
Neuropeptides are molecules similar to a protein, which are created and discharged
by neurons. They regulate the secretory tract and act on neural substrates (Russo
2017). Holmgren and Jensen defined neuropeptide as a peptide discharged by neuron
as a signaling molecule. This signaling molecule will impact other excitable cells as
a modulator or a transmitter (and actually affecting its own cells sometimes)
(Belzung et al. 2006). A considerable point is that neuropeptide is not only just a
peptide existing in neurons but also acts as a medium of communication within the
cells, which means it stimulates other cells through high-affinity binding toward
particular targets (receptors). These molecules are articulated in peripheral nerves
like sensory and motor nerves and also in the central nervous system (CNS).
Rough
endoplasmic Golgi Synapse
Nucleus
reculum apparatus
processing events such as signal peptide cleavage. These are placed in a large dense-
core vesicles (LDCVs). Subsequent to the LDCV exocytosis, the re-internalization
of the LDCV components occurs in the component part of the membrane. Thus, no
neuropeptide is re-used in the synapse. Generally, neuropeptides are released at low
cytosolic Ca concentrations, though LDCV exocytosis is usually stimulated by Ca
ions, whereas the same ion may be utilized for exocytosis when derived through
other important sources such as transmembrane internal stores (Fig. 15.1;
Table 15.1).
Along with it, neuropeptide also imposes certain alteration on behavior. The
alteration in the peptide effect behavior is demonstrated by the gene manipulation
experiments either by transgene mutations or viral gene transfer, exhibiting changes
in the behavioral traits which are part of the expression of peptide receptor (Ludwig
2011). This shows that the peptidergic neurons in the brain do not determine the
behavior but the peptide distribution. The human neurological disorder, such as
depression, autism spectrum disorder, schizophrenia, and social anxiety, has been
associated with vasopressin and oxytocin. In addition, there are neuropeptides
involved in appetite regulation, mood disorder, and libido disorder, which are
included in the peptide-based therapies.
506 N. A. Nimer et al.
In the last 40 years, neuropeptides and their receptors with similarity have been
steadily increasing. At first, the isolation of peptides from the brain and guts can be
observed (e.g., substance P, somatostatin). This can be done by targeted mining in a
particular region that includes orexin in the brain, cortistatin, etc., as well as G
protein-coupled receptors that are deorphanized (GPCRs including ghrelin receptors
and orexin). Moreover, the accomplishment of the Human Genome Project can also
be seen (Schalla and Stengel 2018). The distribution of neuropeptides is being
regionally restricted in the central and peripheral nervous system. The signaling of
neuropeptides and their receptors is more distinct in terms of spatial than signaling
with the classical as well as low-molecular-weight neurotransmitters. These
neurotransmitters seemed to be widely expressed due to the assumption that drugs
that act on neuropeptide receptors possess more selective actions pharmacologically
with a few side effects. However, drugs reacting on glutamatergic, cholinergic,
monoaminergic, and GABAergic systems are less selective pharmacologically.
The functions of neuropeptides include neurotransmitting and acting as growth
factors. They are observed in the hormones found in the endocrine system such as
glial cells; they also act as the messengers in the immune system. When the nervous
system faces any complications (e.g., by stress, injury, or drug abuse), these
neuropeptides seemed to be important. This leads to a massive number of
opportunities to produce new drugs to treat disorders belonging to the nervous
system. As a result of this, subtypes of receptor agonists and certain antagonists as
well as the substance P receptor (neurokinin-1) were discovered. This has proved its
clinical efficiency while treating depression and in chemotherapy induction. Addi-
tionally, several other neuropeptides are under clinical trials for different indications
(Hoyer and Bartfai 2012).
These receptors are assumed to contribute to the discovery of potential drugs
linked to various neurological disorders. According to the researchers, there is a dire
need for antiepileptic drugs (AEDs) which act in several ways than the drugs already
present in the market. A large number of AEDs block the sodium channels to act and
also increase the GABAergic transmission. The therapeutic options are also
expanded by a completely new generation of AEDs; however, these are not as
effective as the older drugs (Hanaya and Arita 2016). To these medications, patients
having mesial temporal epilepsy (mTLE) seemed to be the most pharmacoresistant
(Pati and Alexopoulos 2010). With the aim of rectifying the dilemma, finding the
new mechanism of AEDs is not the only need of neuropharmacologists but to also
focus on the information shared on the pathophysiology of epilepsy. Moreover,
several mechanisms can be observed during the complex procedure of
epileptogenesis; therefore, new compounds should be identified ideally that can
target the other pathways simultaneously. While studying the pathogenesis of
epilepsy, a number of neuropeptides including somatostatin, galanin, and neuropep-
tide Y were briefly studied. However, these neuropeptides are poor and are seen as
the main hurdle for developing the brain drug due to their poor ability to penetrate
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 507
the blood-brain barrier (BBB) (Robertson et al. 2011). The discovery of ghrelin in
1999 brought great excitement to all scientists (Chen et al. 2017). It has been
produced both in peripheral and central terms in the field of epilepsy (Fig. 15.2).
While comparing the well-established anticonvulsant neuropeptides, ghrelin is
recognized to possess a large number of advantages. This neuropeptide has
the ability to cross the BBB while also possessing the characteristic to affect the
physiological processes. The physiological procedures range from the
neuroprotective properties and potent anti-inflammatory along with BBB protection
and integrated hippocampal neurogenesis (Portelli et al. 2012). Besides, various
research studies indicated that physiological procedures are affected negatively
during the process of epileptogenesis (Coremans et al. 2010; Vezzani et al. 2011;
Marchi et al. 2012; Parent and Kron 2012; Vezzani et al. 2013). The recent phase III
clinical trial of ghrelin receptor agonist JMV1843, which highlighted the lack of
hormone among humans, seems to be completed, and the drugs were observed to be
well effective. A company Aeterna Zentaris is successfully developing this drug
now. Moreover, through clinical trials, orexin receptor seemed to be developed and
progressed to treat sleeping disorders. Almorexant completed the phase III clinical
trials (Hoever et al. 2012) and serves as unrecognized orexin receptor antagonist
(Owen et al. 2009). The production of this drug has stopped to a greater extent, as
provided through a brief review of data that was achieved through clinical studies.
This further served as an avenue to demonstrate the advantages of almorexant
(Actelion 2011). The next drug includes suvorexant (MK-4305) which has also
accomplished the phase III clinical trials to identify its effects on different sleeping
patterns (Cox et al. 2010). Based on the classical system of neurotransmitter,
majority of the drugs were approved for treating obesity, while the potential ability
of the system which provides important side effects was ignored. Lorcaserin
(Belviq), a 5-HT2C receptor, seems to receive FDA approval recently for treating
508 N. A. Nimer et al.
obesity in selected obese patients. Weight loss can be seen to be greater than 5% by
placebo-subtraction which was achieved by 27.2%. Maintenance of weight loss by
following the same treatment is observed in 68% of patients (Fidler et al. 2011).
However, the impact of lorcaserin cannot be understood completely specifically for
body weight. However, by mediating it with hypothalamic POMC, activation can be
accomplished (Halford et al. 2007). Mild side effects are revealed in the clinical trials
of lorcaserin while treating for a headache (Smith et al. 2010).
Neuropeptides are also recognized as the molecules that are significant in deter-
mining neurological disorders such as epilepsy and depression. The integration of
the peptides as pharmacological components serves as a great proposition given its
decreased level of toxicity related to the functioning of the metabolism, which also
escalates its potency. Reflecting on an in-depth understanding of the neurological
disorder, it has been found that their use in the therapeutic methods has remained
limited particularly for the treatment of clinical problems. The main contributing
factor for their deficiency of delivering adequate results in the clinical treatment is
their intact peptide delivery to particular regions of the brain, which is essential for
treating the neurological disorder. Generally, the delivery of the drug, which is
derived from the peptides, is confined to two factors: the first includes the issues
concerning the general bioavailability, while the second deals with providing infor-
mation regarding blood-brain barriers. Several factors have been recognized in
creating an impact over pharmaceutical bio-presence of the brain. The factors
comprise the distribution, which takes place in the cardiovascular space, its total
volume, the disappearance of the half-life, as well as the drug capacity for causing
the biological effect by reaching the target. All these factors contribute differently to
each of the drugs, and in terms of the neuropharmaceutical peptide, this serves as a
likely problem, which can be associated with the delivery of the drug.
Depending on the distribution of the cardiovascular system, the component can
be divided into three fractions, namely, protein, blood cells, and protein free fraction
(PFF). In the latter category, the existence of peptide drugs is usually achieved when
it is being transported to the brain. The dynamic equilibrium is maintained by these
three components for their peptide distribution. Generally, the core objective of this
equilibrium is to evolve peptide in PFF, as it is in continuous depletion due to the
action of the metabolic enzymes, its excretion as well as its uptake. Moreover, the
cardiovascular compartment is of great importance as numerous
neuropharmaceutical peptide areas are available in the analogous form of the
endogenous peptides along with its capacity to interact with two other carriers
such as proteins and peptidases. Moreover, it must be emphasized that the level,
which these comprise, can be regulated by enzymes, as well as proteins as the change
in the disease takes place along with the variation in species. Insulin-integrate
growth factor (IGF)-1 that is usually comprised of approximately 70 amino acids
is further assimilated into the binding locations found within the plasma; each
location however has its own different constant for binding. One of the
IGF-binding proteins has demonstrated changes due to its IGF level regulation as
well as changing disease state. The efficiency of the peptide is affected by various
endogenous peptides, which are present in the serum as well as in the blood vessels.
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 509
Example includes the peptide of amino peptidase A, which exists in the serum, to
beat the high level in pregnant women than non-pregnant women. The presence of
BBB also sets parameters for the peptide delivery to the brain. It is because it exists
at the brain micro-vascular capillaries at the endothelial cell level. The resistance
exists because of its integration of the increased level of resistance from the
transendothelial electrical resistance, i.e., 2000 cm in comparison with the peripheral
vessels where it is 3–30 cm. Moreover, the number of endothelial cells in BBB is
low, which reduces the vesicular transportation. BBB also causes neuropeptide
degradation due to its inclusion of the proteolysis enzymes encompassing amino
peptidase A, amino peptidase M, and angiotensin-converting enzyme. In addition,
alkaline phosphatase, glutamyl, transpeptidase, and monoamine oxidase enzymes
are also present in it, which increase the micro-vessel level in the brain and may or
may not be expressed in the peripheral vessels at low levels. It also contributes to the
significant role in the brain homeostasis as well as in several transport systems
allowing the entrance of the substance toward the brain, where large substrates are
present in each of these systems inclusive of several peptides, which can be
transported actively.
Peptide holds relatively strong potential for therapeutic regulation for prevention
against gene and treatment, though it remains limited as a drug due to various
obstacles. In addition, the drug formation of peptide specifically developed for the
disorders related to the central nervous system has remained limited, as a result of
BBB. The peptide drug development and its delivery are being enhanced, which
improves the delivery of the drugs. Previously it was not possible, such as the
concept of the particularly directed antibody vectors as well as glycosylation as its
effect is not only confined to the enhancement of the pharmacological profile of the
drugs but also increases the transportation of the BBB to a particular region in the
brain. The drug delivery of the neuro-peptide can be demonstrated by the utilization
of the vector-mediated delivery, which is a promising area for improving the peptide
targeting, along with gene, where the possibility of success remains high. The
application of these methods is of great assistance in chemotherapeutic treatment
of disease related to the chronic CNS such as Parkinson disease, along with the
effective delivery of neuroprotective agents in the case of acute disorders such as
strokes. To enhance the bio-distribution of the brain, glycosylation is found to be an
effective approach, which improves the stability and mitigated clearance as well as
enhance the transport of the BBB. The enhancement of the opioid peptide related to
the analgesia serves as a great area for recognizing the potential engraved in the
glycosylation strategy. The strategy is also reflected as a source, which can improve
the drug administration for a long period of time, for treating depression or patients
suffering from chronic pain. It is projected that peptide-based CNS drug may prove
to be a composite of more similar methods, of which its discovery through more
research and development in this area continues.
510 N. A. Nimer et al.
It is often tricky to classify these types of peptides with the discovery of endocrine
peptides and neuropeptides, as well as their area of production and their target. This
gives rise to various approaches to the classification of neuropeptide; some classifi-
cation of neuropeptide are illustrated below (Table 15.2).
The hypothalamus is a part of the brain containing various neurons responsible for
the release of numerous hormones. It is located just above the brainstem, below the
thalamus. The hypothalamus is present in all vertebrates and in humans, and its size
almost the same as that of an almond. The hypothalamus is involved in various
regulatory mechanisms and other related functions of the autonomic nervous system;
it produces and releases neuropeptides and neurohormones. Due to the presence of
receptors, there is an increased interest in extra-pituitary actions of these
neurohormones in several non-pituitary tissues. Furthermore, the presence of
The pituitary gland is placed beneath the brain, in the midline pocket of fossa, which
is a small space or cavity in the sphenoid bone. This cavity is also called the “sella
turcica” or the “Turkish Chair”; it is a saddle-figured cavity found in the sphenoid
bone of the human skull and also in other Hominidae skulls, which are the great
family of primitive apes, chimpanzees, gorillas, and also orangutans. The pituitary
gland in humans is made up of two lobes, namely, posterior and anterior lobes.
However, the posterior lobe includes one third of the given area, while the anterior
lobe is based on two thirds of the area. The plain radiography has been replaced with
the availability of modern radiological techniques of the sella turcica to examine
hypothalamo-pituitary abnormalities (Tekiner et al. 2015).
The posterior lobe of the pituitary gland is a lump located underneath the
hypothalamus on the bottom of the brain. The neurons present in the hypothalamus
gland are launched directly to the posterior lobe which are associated with the
pituitary gland. Approximately 100,000 axons are projected from the hypophyseal
nerve tract. The nerve terminals and axons from hypothalamic neurons constitute the
posterior lobe of pituitary gland. The hormones stored in the terminals are secreted
by electrical excitation. Moreover, the altered astrocytes which are also called
pituicytes surround the nerve terminals. The modified astrocytes, known as
pituicytes, surround the nerve terminals, which assume to have a significant role in
the local control of hormone release (Nussey and Whitehead 2013). Various
hormones are produced by pituitary cells and secreted into the bloodstream affecting
other organs of the body. It is also referred to as “the master gland” because it
releases various kinds of peptide hormones which regulate the functions of many
other mechanisms.
The pituitary gland is responsible for sensing the needs of the body, where it
sends signals to different organs and glands throughout the body to regulate their
function and maintain an appropriate environment after sensing. The hormones
secreted by pituitary gland act as messengers after being released in the bloodstream.
512 N. A. Nimer et al.
These messengers are responsible for transmitting information from the pituitary
gland to distant cells, regulating their activity. Spontaneous electrical activity is
enough for driving the intracellular calcium concentration exceeding the threshold
for stimulus transcription and stimulus secretion coupling in some cells. The func-
tion of these action potentials is to retain the cells with cytosolic calcium in a
response state near the threshold phase (Stojilkovic et al. 2010). Some of the
pituitary gland hormones have been indicated below with their functions:
15.4.3 Tachykinins
Putative neuropeptides refer to the neuropeptide precursors and encoding genes that
are attained through a biologically active process. Not all members of the cerebellin
family meet the definition of a neuropeptide; in particular, the regulated discharge
has not been recognized for each member of this family. The names of the genes are
thus abbreviated using the standard nomenclature for genes and their given localiza-
tion in the human genome. Neuropeptides and their receptors are concerned in
behaviors, specifically in mammals, which include sleep and feeding. Neuropeptide
functions are yet not understood, despite their clear roles in synaptic behavior and
signaling (Nathoo et al. 2001).
In various cases, tachykinins are extracted in the form of an enzyme from the
existing proteins. This helps in the formation of products that are biologically active
and functional. The neuropeptides can be achieved through the alternate processing
of three significant TAC genes. The genes are further encoded by preprotachykinin
genes, where the preprotachykinin A (TAC1) gene helps in encoding the sequences
of neurokinin A (NKA) and SP. In contrast to the two, the third category (TAC3)
helps in the uniformity of neurokinin B (Kalina et al. 2018). Besides, α-amidation is
generated through the bio-activation of SP that is integrated through α-amidating
monooxygenase and peptidylglycine. The α-amidation is functionally limited to
provide α-amidated peptides. Other enzymes such as the angiotensin and endopepti-
dase help in altering or minimizing the impact of α-amidation (Scholzen and Luger
2004; Mendlewicz et al. 2005).
The degradation and synthesis of substance P is shown in Fig. 15.3. Plasma levels
of SP in humans ranges between 30 pg/ml and 500 pg/ml (Bondy et al. 2003; Lee
et al. 1997; Reynolds et al. 1988).
Neurokinin A
TAC1
Substance P
(Inactive)
Peptidylglycine α-amidating monooxygenase
Substance P
(NK1R)
(Active)
Angiotensin-converting enzyme Neutral endopeptidase
Carboxyl Terminal Enzyme C4
Degradation Amino
Terminal
Substance P binds to NK1R, NK2R, and NK3R receptors in target tissues (Yin et al.
2018), having the highest affinity to NK1R; the biological effects of SP are thus
mainly mediated through this receptor (Chi et al. 2018).
There are two isoforms of NK1R, which are derived from differences in the
tachykinin receptor gene and are different depending on the cytoplasmic tails of
the C-terminal. The number of amino acids is also different, as one of the categories
consists of 407 amino acids. The second, i.e., the truncated receptor, is based on
516 N. A. Nimer et al.
311 receptors. The bonding power of SP is ten times higher for the receptor available
in full length in comparison to the truncated receptor (Chi et al. 2018). The isoforms
are different depending on their impact over the metabolic functions of cell and thus
result in separate pathways (Steinhoff et al. 2014; Lai et al. 2008). The long isoform
takes just 1–2 min to phosphorylate extracellular-regulated protein kinases, while
this cellular response is not observed until considerable time, at least 20 min, has
elapsed after exposure to SP in cells expressing truncated NK1R (Chi et al. 2018).
The mediation of complete NK1R helps in cyclic adenosine monophosphate
(cAMP), while Ca2+ helps in the formation of binding protein. It further helps in
increasing the mechanism of gene transmission. The independent mechanism of Ca2+
helps in signaling the truncated NK1R of the C-terminal (Lai et al. 2008).
The formation of the transmembrane domains is initiated through a NK1R
polypeptide chain that is functional by seven times providing seven different trans-
membrane domains. It is further connected to extracellular and intracellular loops
(i.e., EL1, EL2, and EL2 and C1, C2, C3). However, the extracellular terminal is
found in amino (N) terminal of the receptor, while intracellular terminal is located at
the carboxyl (C) terminal of the receptor. G protein interacts with cytoplasmic
regions of the receptor, especially around loop C3, connecting the fifth and sixth
transmembrane domains. The binding sites of antagonists and agonist are found in
the second and third transmembrane domains (Chi et al. 2018). Figure 15.5
illustrates the layout of full-length NK1R.
Different types of G proteins (Gαs, Gαi, Gα12/13, Gα0, Gαq) that are attached to the
receptor are functional in demonstrating the intracellular pathways through
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 517
include acute antral ulcerations of the stomach tissue. The increasing amount of
SP-immunoreactive Auerbach’s response in the porcine stomach helps in proving
the significant value of the given neurons for neuroplasticity, since the inflammatory
problems are highly responsive to ulcer diseases. This, however, promoted the
indirect functioning of SP in the gastric nerve pathways and submucosal neurons
to provide a massive supply of pyloric sphincters in various animals with antral
ulcerations. This indicates that majority of the issues in the given regions are often
connected to inefficient functioning of SP providing no significant contribution in
regulating the myenteric neurons of the stomach (Zalecki 2019).
The crystal structure of human NK1R bound to a high-affinity antagonist reveals the
molecular basis of antagonist interactions and offers information toward understand-
ing ligand binding selectivity for this pharmacologically therapeutically important
family of GPCRs (Yin et al. 2018). Recently, SP and NK1R have gained attention
based on the development of complex NK1R antagonists that have many promising
therapeutic indications for issues such as depression, pain, and emesis (Campbell
et al. 2006). It has been shown that NK1R is significant in creating the impact on
different activities of the brain including emesis centers. Aprepitant which is an oral
antagonist is proved to be an impactful agent in preventing acute and delayed
chemotherapy-induced nausea and vomiting (Manak et al. 2010; Muñoz and
Coveñas 2013; Schank and Heilig 2017; Yin et al. 2018). Maximum exposure of
aprepitant is important in creating a significant impact over the physiological
activities that include the unbalance metabolism of cholesterol and cell trafficking,
inflammation, and apoptosis. Plasma levels of SP decrease when treated with
aprepitant due to a significant impact of over 176 plasma proteins and other
pathways that are important for metabolisms including lipid metabolism and inflam-
mation. The agent is further related to a significant increase in HDL cholesterol. Also
significant impact is provided to different metabolic and hematologic markers,
which are further helpful in restoring the baseline levels after 30 days of aprepitant
treatment (Spitsin et al. 2017).
The ability of aprepitant treatment is significant in coping with the blood-brain
complexities; its approval as drug to reduce chemotherapy complexities provided by
FDA helps in giving it a significant status in treating infections related to human
immunodeficiency virus (Manak et al. 2010). Cases that include the NK1R as an
antagonist help in suppressing cholesterol that in return provides significant
macrophages. This helps in protecting the metabolism from atherosclerosis (Spitsin
et al. 2017).
Other oral NK1R antagonists, including netupitant and rolapitant, cross the blood-
brain barrier and bind brain NK1R. They are highly selective and have no affinity for
serotonin, dopamine, or corticosteroid receptors. However, netupitant (300 mg) is
available only in a combination formulation with palonosetron (0.5 mg).
Fosaprepitant is an intravenous pro-drug of aprepitant that is converted within
30 min after infusion (McQuaid 2018). These NK1R antagonists are safe and well
tolerated, and most adverse events are minor and well tolerated, such as headaches
(Muñoz and Coveñas 2013), fatigue, and dizziness (McQuaid 2018).
An increasing number of studies on NK1R antagonist have been conducted which
focuses on psychiatric processes and disorders like depression, stress, and anxiety.
522 N. A. Nimer et al.
The clinical developments, however, indicated the failure of the given treatment.
Several studies in the given framework have indicated the importance of taking
NK1R drug as it has impact on the treatment of stress-related problems. Despite
greater efforts, studies remain unsuccessful in increasing the usage of drugs among
the given populations. The study further demonstrated that the inefficiency of NK1R
antagonists in treating alcohol addictions is due to the insufficient presence of
receptors. As a result of this, several developments are being made on the functions
of NK1R to complete the target of providing new pharmacotherapeutics in treating
addiction problems (Schank and Heilig 2017).
With regard to pain transition, SP receptor antagonists do not block responses to
certain types of pain, though they can modify some responses where glutamate,
which is often extracted through SP, provides a greater significance in eradicating
pain (Gray 2018). As a therapeutic agent for treating epilepsy, NK1R antagonists
reduce kainic acid-induced seizure activity, though their therapeutic potential for
treating epilepsy has not yet been fully exploited (Wang et al. 2017; Chi et al. 2018).
Additionally, human NK1R modulators have shown promise in clinical trials for
migraine and depression (Yin et al. 2018). In particular, there is a greater similarity
found between SP receptor antagonist MK-86 and paroxetine due to their anxiolytic
and anti-depressant effect that is usually attained in treating patients with unipolar
disorder (Kramer et al. 1998). The models related to the chronic obstructive pulmo-
nary disease have significantly provided decrease in inflammation in the respiratory
tract when treated through tachykinin receptors (De Swert et al. 2009). The insuffi-
cient evidence provided in this regard demonstrates that the antagonists are impor-
tant to minimize the airway responses while improving the function among patients
with asthma (Ramalho et al. 2011).
In terms of addressing tumor genesis, there is also evidence to indicate that NK1R
antagonists may have value in treating patients with colorectal adenomas (Gao and
Wang 2017).
the endocrine cells, the digestive tract, and the central nervous system. VIP is further
important as it regulates secretion and blood flow in both the intestine and pancreas.
This helps in increasing the gastrointestinal motility due to the display of similar
effects in urological, respiratory, and cardiovascular system. Reports provided for
this neuropeptide have recently identified a wider range of activities, highlighting its
neuroprotective and immunological effects (Dickson and Finlayson 2009; Hisato
Igarashi et al. 2011). The ligands along with the Class II G protein-coupled receptors
have a great therapeutic potential, as suggested by clinical data along with similar
investigations (Chapter et al. 2010). Still, it is important to analyze the developments
of applications in various ongoing disease and pathological disorders, along with the
structural and functional value of neuropeptides and its receptors (Hisato Igarashi
et al. 2011). The existing information related to VIP and its receptors involves
several pharmacological and clinical findings that are relevant to the existing
developments in the given problem. Therefore, the following sections provide a
detailed description about the given idea.
VIP
PACAP
1
2
Ser
His Nh2 N-terminus
3
Asp
4
Ala 5
Val 6
Phe β-bends
Thr
8
21
Asp 22
9 Lys 20
Tyr
Asp 23 Lys
19
10
Leu Val
Tyr 24
Asn
11 Thr 18 Ala
12 17
25 Arg
α-helix
Ser Met
13 16
Leu 14 15
Ile Gln
27 Asn
28
et al. 2009; White et al. 2010). Figure 15.6 demonstrates the high degree of the
amino acid sequences homology shared by both VIP and PACAP peptides.
Analyses of VIP using nuclear magnetic resonance spectroscopy (NMR)
provided a helical conformation with α-helix and two β-bends provided at the
given residues of 11–26 and 2–5 and 1–10, respectively. The helical conformation
is located at the N-terminus as indicated in Fig. 15.7. The two domains including the
C-terminal and N-terminal are important, as they play a significant role in managing
bio-activity and receptor recognition (Igarashi et al. 2011).
In 1995, cloning and mapping of a VIP gene was identified in humans and is
placed at chromosome 6q25. The structural formation of a human VIP gene is based
on 7 exons that are incorporated with 6 introns along with the spans of 9 kb. The
gene is further significant in providing the two peptides that are biologically active
and highly functional. The formation occurred through the incorporation of
170 amino acid preproteins tailored in a proteolytical manner. The two resulting
peptides are known as peptide histidine methionine (PHM) and VIP (Dickson and
Finlayson 2009; Dorsam et al. 2011).
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 525
VIP is based on two definite receptors including VPAC1 and VPAC2 that are
mutually connected through PACAP and VIP to a certain extent. The given receptors
are important and thus belong to a class B group of the G protein that are further
coupled through seven different transmembrane receptors. It further consists of
approximately 437–459 residues of amino acids followed by a long chain of
N-terminal (Laburthe et al. 2007; Vaudry et al. 2009). The availability of
N-terminal with a structural representation of N-cap indicates that it is highly
involved in the stimulation of receptors and could be implicated in providing
important developments in terms of treatment design that targets different
membranes of B GPCRs and VPAC receptors (Neumann et al. 2008). However,
the α-helical conformation is usually indicated through the binding of peptides along
with certain specific receptors (Laburthe et al. 2007; Vaudry et al. 2009).
The examination regarding the distribution patterns of VPAC1 and VPAC2 is
provided in abundance. The allocation of VPAC1 specific to certain regions of the
central nervous system includes the piriform cortex, putamen, choroid plexus, lateral
amygdaloid nucleus, etc. Besides, the functionality of VPAC1 is provided in
kidneys, small intestine, liver, mucosa of the stomach, prostate gland, spleen,
lymphocytes, mammary glands, and many other tissues. Contrary to this, VPAC2
is extensively found in smooth muscles of various organs and vascular walls that
specifically include the large intestinal mucosa, heart, retina, pancreas, adrenal
medulla, alveolar epithelium, testis, and adipose tissue. VPAC2 when functioning
in the central nervous system is usually found in the cerebral cortex, amygdala,
hippocampus, olfactory brain, paraventricular cortex, and thalamus (Hisato Igarashi
et al. 2011).
VIP and PACAP receptors are stimulated by several endogenous peptides that
vary in their affinities such as VIP, PACAP-38, PACAP-27, peptide histidine
methionine amide (PHM), peptide histidine isoleucine amide (PHI), and peptide
histidine valine (PHV). Similar binding strengths have been identified in VIP and
PACAP by both receptors. However, PACAP27 and PACAP38 with 1000-fold
reflected maximum affinity in comparison to VIP that serves as the antagonist of
isoforms found in the PAC1 receptor (Henning 2013). VIP and PACAP are impor-
tant in maintaining the physiological mechanism of several systems that contribute
to important pharmacological effects in the human body as described in the sections
below.
The peripheral and central nervous systems and gastrointestinal tract are comprised
of VIP neuropeptides that are found in abundance. The given neuropeptide is crucial
in functioning as a neurotransmitter and neuromodulator for different physiological
functions including ion transportation, neuronal functionality, and mucosal
secretions. It further helps in vasodilation in endocrine, cardiovascular, pancreatic,
526 N. A. Nimer et al.
• Relaxation of blood vessels and smooth muscles by inducing the release of nitric
oxide (NO)
• Enhancement of electrolytes and diaresis by increasing urine volume and enhanc-
ing fractional excretion of sodium, chloride, and potassium
• Regulating the neuroendocrine and endocrine functions
hormones from the pancreas. It is further important in increasing the flow of blood
through vasodilation (Dickson and Behrman 2013; Chandra and Liddle 2015). VIP
and its close relative PACAP also promote insulin and glucagon release through
several ways, such as the secretion of insulin through cAMP coupling metabolism,
extracellular signal-regulated kinase (ERK) pathway along with signaling of P13K,
β-adrenoceptors. Others include the mobilization of Ca2+ that is integrated through
the intracellular stores of the gastrin-releasing peptide (Röder et al. 2016).
Studies further indicated that the anterior pituitary and hypothalamus are used to
secrete and synthesize VIP. Also, it helps in making accurate regulations of the
pituitary functions including FSH, LH, TSH, and prolactin, T3 and T4 thyroid, and
gonadal and adrenal hormones, in response to acute inflammation provided through
LPS (Bik et al. 2004). Immunoreactive VIP that is found in the hypothalamus and
pituitary usually increases due to the treatment provided through estrogen and
adrenalectomy. Moreover, the immunoreactive VIP decreases followed by the
hyperprolactinemic states. During the duration of sexual maturation and lactation,
the levels of VIP mRNA in the hypothalamus are significantly high. Results however
provided the physiological role of pituitary and hypothalamic VIP gene expression
in relation to its fundamental function as neuroendocrine hormone. Moreover, VIP
stimulates the release of luteotropic hormone called pituitary prolactin (PRL) from
the pituitary at different levels that can easily be obtained from the hypophyseal-
portal blood, while including other pituitary functions, such as release of growth
hormone, ACTH, and vasopressin, and stimulates the release of catecholamine from
the adrenal medulla.
The control of the PRL secretion is integrated through the endogenous VIP found
at multiple locations. This further helps in maintaining firm interaction with other
moderators of the PRL secretion that include prostaglandins, cholecystokinin, sero-
tonin, and oxytocin (Chaiseha et al. 2010; Blanco et al. 2013).
VIP serves as the fundamental regulator of neuropeptide that is extensively
distributed in both peripheral and central nervous system. The representation of
the significant type of endogenous sponsors to the formation and regulation of the
immune variances in various CNS immune organ, further helps in controlling acute
inflammation in various peripheral immune organs (Ganea et al. 2014). Provides an
important recognition to VIP in terms of serving as a valuable anti-inflammatory
factor in adaptive and innate immunity. The production of pro-inflammatory
cytokines and chemokines from dendritic cells, microglia, and macrophages is
integrated through VIP. The process is highly common in innate immunity. Also,
VIP diminishes the expression of costimulatory molecules on the antigen-presenting
cells and consequently decreases the activation of T cells that are highly specific to
antigens. In the case of adaptive immunity, VIP promotes the responses provided by
Th2 and helps in reducing the Th1 type of pro-inflammatory responses (González-
Rey et al. 2004).
VIP is significantly functional in the homeostasis of the respiratory system and
employs a potent vasodilatory and bronchodilatory consequences in the system. It
further encourages the secretion of mucous that is headed through the tracheobron-
chial submucosal glands. Through several clinical trials and research studies, it can
528 N. A. Nimer et al.
Recently, in vivo test was used to test the interneurons that function through VIP
and are mostly found in post-natal development of the cortical circuits. The removal
of the early postnatal ErbB4 helped in dysregulation of the VIP interneurons. In
some cases, it appears during the growth period which is visible in creating
digressions in their functioning. It further leads to high dysregulations in the
relationship between cortical, temporal, and state dependence. The data however
provides valuable insight about the function of VIP interneurons in the formation of
cortical circuit and is also important in creating a significant impact over the
pathophysiology of neurodevelopmental disorders (Batista-Brito et al. 2017).
Vasoactive intestinal peptide belongs to the superfamily of various peptides that are
structurally related and thus includes growth hormone-releasing hormones, secretin,
gastric inhibitory peptide, glucagon, and glucagon-related peptides.
The peptide is important as it exerts its action through VPAC1 and VPAC2
secretin receptor-like. Progress in characterizing the functions of these receptors
has been delayed due to the insufficient availability of drugs. Literatures identify
several antagonists and agonists related to the VIP receptors. [Ala11,22,28] VIP and
[Lys15, Arg16, Leu27] VIP (1–7)/GRF (8–27)-NH2 are certain agonists of the VPAC1
receptor and PG 97–269 is a selective antagonist. The most selective identified
VPAC2 agonist is Ro 25–1392; but on the other hand, myristoylation provides a
-K-K-G-G-T sequence of the amino-terminus for VIP (1–26) [K (12)]. It further
provides the extended carboxyl-terminal of Ro 25–1553 that is a specific antagonist
of VPAC2 receptor antagonist. The molecule provides a VPAC2 receptor antagonist
that is found in humans and is widely present (Moreno et al. 2000; Dickson et al.
2006; Harmar et al. 2012).
Cardiopulmonary system highly reflects the activities of VIP through various
pharmacological functioning that includes; anti-inflammatory actions of, potent
improved blood circulation within the region of lungs and heart, potent airway and
dilatory actions, etc. (Wu et al. 2011). Since VIP functions as a neurotransmitter, a
significant impact is created in the form of vasodilatory and potent bronchodilatory
functioning. Also, it helps in the secretion of mucus through submucosal glands. VIP
functions as immunomodulator, for instance, in suppressing the humoral immune
response, inhibition of bronchial and vascular inhibition, and diminishing the
extracts of cigarette smoke resulting in the loss of L2 alveolar cells. Recent investi-
gation indicates a potential therapeutic role of a novel stabilized inhaled VIP agonist
with minimized side effects dealing with a variety of lung diseases such as pulmo-
nary hypertension, cystic fibrosis, asthma, chronic obstructive pulmonary diseases,
etc. (Mathioudakis et al. 2013).
VIP plays a fundamental role followed by various functions such as the regulation
and control of timing of circadian rhythms, memory, and learning along with various
stress responses. The present study relates VPAC2 receptor in providing vulnerabil-
ity to schizophrenia. VIP, in various peripheral regions, plays a significant role in
530 N. A. Nimer et al.
neurodegeneration over time. In a recent study, VIP was shown to effectively limit
injurious stimulation of Aβ-concentrated microglia and helps in discharging the
neurotoxins including tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and nitric
oxide (NO). Thus, VIP is crucial in preventing the neuronal cell death leading toward
AD pathology in the brain (Delgado et al. 2008; Querfurth and LaFerla 2010).
Publications have resulted in providing significant relationship between
variations of gene encoding including schizophrenia susceptibility and VPAC2
receptor. These results have generated some enthusiasm in the field of new antipsy-
chotic drug developments (Levinson et al. 2011).
The suppression of Th1 immune response along with the activation of T cells is
integrated through VIP, where it serves as the anti-inflammatory mediator to provide
immune balance. In the present era, VIP acts as a therapeutic agent for various
autoimmune diseases such as ulcerative colitis, multiple sclerosis, and rheumatoid
arthritis (Pezzilli 2006).
Current literature reviews show that vasoactive intestinal peptide receptors
(VIPRs) are over functional in some malignant tumors. Also, they provided infor-
mation related to the progression of several malignancies. The analogs of the agents
are labeled through radionuclide and are used for imaging of tumor receptor.
The images are important in visualizing the VIPR surface of protein functioning.
The images were visualized via in vivo test that provides information regarding the
properties of molecular drugs when used for treating tumor. Besides, the relationship
between VIPRs and angiogenesis and malignant transformation is important in the
treatment of cancer (Bo Tang et al. 2014).
The function of VIP specifically in the gastrointestinal tract indicates that
neurotransmitters and its receptors help in dealing with several disorders related to
the secretion of gastric acid along with their impact on gastrointestinal motility
including functional bowel syndrome (Mawe and Hoffman 2013).
Deficiency of nutrients (or caloric restriction) can accelerate the orexigenic peptide
neuropeptide Y (NPY) and autophagy in cortical and hypothalamic neurons
(Catalani et al. 2017). The NPY plays a very essential role in various physiological
functions like energy homeostasis, cognition, and neuroprotection and neurogenesis,
intake of food, stress response, and circadian rhythm; therefore, it serves as an
important neuropeptide that is widely present in the brain (Diaz-delCastillo et al.
2018).
A distinguished hypothalamic neural cell in rat and hypothalamic neuronal cell
line in mouse enhance the flux of neuronal autophagy, which is exhibited by the
study of LC3-II turnover, the rise in the number of autolysosomes and
autophagosomes, and decline in the amount of p62. This impact is exercised by
the triggering of Y1 and Y5 receptors. The protein kinase stimulation such as PKA,
532 N. A. Nimer et al.
PI3K, and ERK1/2-MAPK by NPY results in the signaling of track linked with the
induction of autophagy. By in vivo over manifestation of NPY in the arcuate
nucleus, the flux of NPY-induced stimulation was verified in the hypothalamus of
mice. Furthermore, in cortical neurons of rat, the autophagy is accelerated by NPY
(i.e., the decline of p62 expression and growth of LC3-II) possibly by the prevention
of mTOR activity. In mice, which are nourished with a diet containing a high level of
fats, the removal of AMPK activity within the arcuate nucleus of hypothalamus
reduced NPY expression and autophagy, hence decreasing body weight and intake
of food as a result. Therefore, in the hypothalamic cell lines, increased levels of
NPY, the stimulation of AMP-activated protein kinase, and regulating of mTOR
signaling result in the induction of autophagy. In both, the NPY levels and
autophagy keep declining with age; research is being carried out to enhance the
level of NPY and increase autophagy including caloric restriction, as a result of
which it was recommended to develop defending effects that postpone deficiencies
related to prolonged existence. By regulating hypothalamic autophagy, one might
also be able to eliminate the prospects of obesity and other metabolic disorders
associated with aging.
Lastly, the NPY exercises a neuroprotective effect on the cerebellum and striatum
of spinocerebellar ataxia type 3 of two mouse models, which is a disorder
symbolized by autophagy defects. Therefore, it was recommended that this action
might be associated with the stimulation of the clearance process of protein like
autophagy; however, more data is required to support this hypothesis or assumption.
Generally, the NPY ability to prolong neuro-degeneration by activation of
autophagy as an approach to a free abnormal and misfolded protein that results in
neurodegenerative disease is necessary to be studied in detail.
Neuropeptide Y (NPY) was first discovered in 1982 and was found in the porcine
brain. The information suggested that formation of neuropeptide Y is possible
through the amalgamation of 36 amino peptides and thus is generally functional in
the peripheral nervous system (PNS) and central nervous system (CNS) of
mammals. This neuropeptide comprises pancreatic polypeptide (PP) and peptide
YY (PYY) as it shares its amino acid sequence homology in 70% and 50%,
respectively. The similarity of the sequence in terms of their molecules represents
them in a U-shaped molecule, which is formed in a structure similar to the hairpin,
along with a tertiary structure comprising of an amphiphilic α-helix, which are
joined by the β-turn with a type II helix and similar to polypro line. This structure
of the neuropeptide represents the tridimensional structure, which constitutes peptide
affinity and efficacy connecting them to their several receptors. The NPY biosynthe-
sis translates the molecule of the pre-pro-NPY in which translocation occurs directly
with the endoplasmic reticulum, where the mitigation of the peptide takes place.
Through the prohormone convertases, cleaving of the pro-NPY precursor takes place
in the NPY along with its C-terminal NPY peptide (CPN), where the NPY peptide
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 533
goes through two truncations, i.e., through a peptidyl glycine alpha-amidating mono
oxygenase and a carboxypeptidase for the NPY maturation as well as its activation.
The amidation of the C-terminal serves as the final characterization of the molecule,
which impedes the degradation and takes place through the action of the
carboxypeptidases. With NPY immense consistent distribution of the CNS and
PNS, there are several biological impacts as well as brain disorder, which encompass
disorder like obesity, anorexia, depression, drug addiction, itching, pain, and energy
homeostasis (Loh et al. 2015; Gonçalves et al. 2015; Farzi et al. 2015; Bourane et al.
2015; Arcourt et al. 2017).
related to the spinal cord in the dorsal horn and encompasses the likely targets for the
drug in the treatment of chronic pain.
In recent times, the association of neuropeptide tyrosine (NPY) has been found to be
in neurological responses related to ethanol and drug abuse, which is highlighted
from genetic, pharmacological, and molecular indication (Ciafrè et al. 2016). The
pharmacology of neuropeptide tyrosine has initially been examined from the periph-
eral sympathetic control of the neurons related to the blood vessels, spleen, heart, as
well as vas deferens. The release of the NPY largely takes place when the stimulation
constitutes of high frequency or activation of the strong sympathetic reflex. NPY has
a particular receptor mechanism, which is present at the pre-junction level as well as
post-junction levels, where the existence of an enormous amidated C-terminal
portion of NPY is required to bind the receptor and results in vasoconstrictor effects
as well as the inhibition of cyclic AMP formation. Moreover, the release of the NPY
is also impacted by various pharmacological agents encompassing clonidine, gua-
nethidine, yohimbine, nicotine, angiotensin, angiotensin II, and desipramine.
Among the NPY receptors, the Y5 receptor constitutes the pharmacological profile
particularly for the peripheral tissues, though its mRNA expression is greatly
confined to the CNS (Dan Larhammar et al. 2015).
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 535
The fragments of the NPY C-terminal serve as the source, which characterizes the
NPY receptors. It is due to its NPY and PYY fragment imitation of the responses
which are given by the NPY such as in the twitch responses of the pre-junctional
inhibition as it occurred in rat vas deferens; however, they are not observed in
vasoconstriction of guinea pig iliac vein. In the context of the NPY receptor, it has
been suggested that Y1 are the receptors that are activated by holopeptides, whereas
Y2 are the receptors that are activated by the fragment of the C-terminal and
holopeptides. Although the synthesis of the various C-terminal fragments occurs,
the NPY and PYY of the 3–36, 13–36, and 18–36 fragments are often utilized
irrespective of their obvious benefit between these fragments. The discrimination of
the receptors such as Y1 and Y2 is done using the NPY C-terminal fragment, though
these are not only confined to the Y2 selection as Y5 receptors can also be activated
using the same concentration. It should be noted that the selection of the receptors is
based on the difference between the availability of tools and the substantial differ-
ence concerning the affinity of the receptor in relation to their cognate ligands which
prevails between the species. The recognition of the receptor is based on the various
combination of the complementary agents which are used. The application of this
condition is more momentous for in vivo, whereas the less characterization of the
compounds occurs in contrast to in vitro.
which causes mediating influence on the spinal as well as supraspinal level (Taylor
et al. 2014). In the chronic pain modulation, the role of NPY along with its Y1R and
Y2R receptors is found to be crucial, where the NPY receptors can also be regarded
as a target for a drug where they are used for devising new drug design. However,
various problems exist, which are difficult to overcome with the use of this targeted
treatment. The route of administration of these receptors, such as the NPY agonists
which are peripherally applied, results in the behavior that emerges due to a halt, and
it appears that the analgesic effect occurs only when the peptide or drug formulation
takes place following the blood-brain barrier (BBB). However, the lumbar intrathe-
cal drug delivery is often found to be linked with some patients’ sort of discomfort
and inconvenience, which can be mitigated through increasing the treatment effi-
ciency as well as persistence of its effects, which are still lacking. Another difficulty
associated with the determination of the severe pain in NPY agonist when found in
clinical practices is linked with the deficiency of the non-peptidic small molecule of
the NPY receptor agonists (Mittapalli and Roberts 2014). To treat these cases,
different strategies are adopted such as targeted agonist delivery by using the therapy
related to the nanoparticles or gene (Chandrasekaran 2013).
Huang et al. (2006) initially discovered calcitonin (CT) which is a kind of polypep-
tide hormone (Masi and Brandi 2007). This identification occurred when the regu-
latory hormone for calcium level was being discovered. This peptide is able to
reduce the blood calcium level through the direct inhibition of the mediation of the
bone resorption, which also improves the kidney exertion of calcium. There are
32 amino acid residues present in the single chain of human CT peptide, which has
its molecular mass as 3418 Da. The cysteines are linked through the disulfide bridge,
which connects them from positions 1 to 7 to form a ring structure that includes
seven amino acids located at the amino terminus. Moreover, the CT precursor,
pre-procalcitonin (PreProCT), withholds 141 amino acids (Fig. 15.8).
The stem cells from neural crest as well as the rostral are transported into the
ultimo-branchial glands of lower vertebrate animals and into the para-follicular cells
in humans. The C cells found in the ultimo-branchial glands of lower vertebrates and
in the thyroid gland of mammals produce calcitonin (CT) and release them into
circulation, which works in a similar manner to other hormones in the body. Through
this advance movement, it is assumed that concentration of C cells is required in
ultimo-branchial body and thyroid gland (Johansson et al. 2015). However, it is
often noticed that some C cells might not be able to make their way forward toward
the thyroid and end up in the extra-thyroidal tissues during their forward movement
(Giovanella et al. 2013). Calcitonin is considered as endogenous controller of
calcium homeostasis and protects the skeleton especially in “calcium stresses” by
acting primarily on the bone. Calcitonin also acts directly on gastrointestinal and
kidney secretion activity by creating an indirect impact on the central nervous system
(CNS) to lessen the pain. Studies have found that the role of calcitonin is not limited
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 537
5’ I II III IV V VI 3’
0
0
0 TRANSCRIPTION, MATURATION
0 SPLICING AND POLYADENYLATION
0
0
0
Calcitonin I precursor
77777 Calcitonin II precursor CGRP I- precursor
I II III IV V VI
to intrinsic analgesic effects on CNS and thus creates a significant impact over
various neuronal activities, like moderating them directly on their existing location
or indirectly moderating them by a process which is yet to be explained on other
locations. Presently, the main indications for using calcitonin in therapies are
disorders, which include high-bone turnover osteoporosis, acute pancreatitis, osteo-
porosis linked to pain or metastases of bone, hypercalcemia, Sudeck’s atrophy, and
Paget’s disease (osteitis deformans). Many kinds of calcitonin are commonly con-
sumed such as synthetic human calcitonin (SCT), analog (Elcatonin), synthetic
salmon calcitonin (SCT) also known as salcatonin, natural porcine calcitonin
(PCT), and synthetic eel calcitonin (ECT). One of the problems with its treatment
is that the only imaginable way for its administration is through an injection,
although other ways for its dosage are being explored.
538 N. A. Nimer et al.
During the evolution, calcitonin has been well preserved. Calcitonin from 9 distinc-
tive species has been recognized with the identification of 12 different sequences.
The remains of six unchangeable amino acids are found at the amino terminal, while
two others are obtained through the end of the carboxyl terminal of the peptide
molecule.
Moreover, at C-terminal, all the calcitonins have 1–7 disulfide bridge and proline
amide. For the osteoclast inhibitory functions and for hypocalcemic bioactivity, all
the 32 amino acids are needed. The substitution of certain amino acids during the
synchronization of synthetic calcitonin (like synthetic eel-CT) helps in increasing
their lifetime to a certain extent by struggling against degradation, hence improving
their circulatory half-life and its biological function. The method involves certain
risks as it may result in allergenicity. Calcitonin is prepared as a precursor molecule.
Before the release of the fully developed form of biologically active calcitonin,
various C-terminal amidation and post-translational changes, such as cleavage, take
place. Glucagons, theophylline, cholecystokinin, dibutyryl cyclic AMP, gastrin, and
cations like Mg2+ and Ca2+ accelerate the secretion of calcitonin from C cells. The
impact of calcitonin over osteoclasts can be represented through dibutyryl cyclic
AMP. However, the dose dependency to eliminate the cyclic AMP is integrated
through calcitonin.
The quantification of plasma i-CT levels (i.e., for hypertension) is being utilized for
diagnostic and inspection tests for C cell hyperplasia for the diagnosis of
pre-malignant and malignant disorders that are related to C cell (i.e., patients’ families
suffering through medullary thyroid carcinoma (MCT)) (Wimalawansa 2010). Cur-
rently, there is the availability of various diagnostic-stimulation tests for the diagnosis
of MTC like an infusion of pentagastrin and injecting calcium (Fig. 15.9).
Fig. 15.9 Human calcitonin amino acid sequence (Source: Wimalawansa 2010)
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 539
Calcitonin has been observed to play a significant role in the treatment of pathogenic
diseases. The effectiveness of calcitonin is well-established for providing treatment
to patients who are suffering from Paget disease. The raised Pagetic bone turnover
indices are decreased when the calcitonin is administered along with the subcutane-
ous injection of about 50% (Pondel 2000).
The symptoms related to the Paget disease are also reduced with the use of CT
which encompass disease comprising pain in the bones and complications related to
the neurology comprising nerve root compression, headache, and spinal stenosis.
Along with it, osteoporosis is characterized as the systemic skeletal disease which
comprises low bone mass as well as bone tissue deterioration which result in fragility
in the bone along with fracture susceptibility. The decrease in the levels of estrogen
which takes place at menopause results in increase resorption of the osteoclastic
540 N. A. Nimer et al.
Man Rat S-1 S-2 S-3 Eel *Chiek Bov Pore Ovi *Man2
1 Cys - - - - - - - - - Tyr
2 Gly - Ser Ser Ser Ser Ala Ser Ser Ser Ser
3 Asn - - - - - Ser - - - -
4 Leu - - - - - - - - - -
5 Ser - - - - - - - - - -
6 Thr - - - - - - - - - -
7 Cys - - - - - - - - - -
8 Met - Val - Val Val Val Val Val Val Leu
9 Leu - - - - - - - - - Gln
10 Gly - - - - - - Ser Ser Ser -
11 Thr - Lys Lys Lys Lys Lys Ala Ala Ala -
12 Tyr - Leu Leu Leu Leu Leu - - - -
13 Thr - Ser Ser Ser Ser Ser Trp Trp Trp Leu
14 Gln - - - - - - Lys Arg Lys -
15 Asp - Glu - - Glu Glu - Asn - Tyr
16 Phe Leu Leu Leu Leu Leu Leu Leu Leu Leu Leu
17 Asn - His His His His His - - - Lys
18 Lys - - - - - - Asn Asn Asn Asn
19 Phe - Leu Leu Leu Leu Leu Tyr - Tyr -
20 His - Gln Gln Gln Gln Gln - - - -
21 Thr - - - - - - Arg Arg Arg Met
22 Phe - Tyr - - Tyr Tyr - - Tyr -
23 Pro - - - - - - Ser Ser Ser -
24 Gln - Arg Arg Arg Arg Arg Gly Gly Gly Gly
25 Thr - - - - - - Met Met Met Ile
26 Ala Ser Asn Asn Asn Asp Asp Gly Gly Gly Asn
27 Ile - Thr Thr Thr Val Val Phe Phe Phe Phe
28 Gly - - - - - - - - - -
29 Val - Ser Ala Ala Ala Ala Pro Pro Pro Pro
30 Gly - - - - - Glu Glu Glu Glu Gln
31 Ala - Thr Val Val Thr Thr Thr Thr Thr Ile
32 Pro - - - - - - - - - -
bone, up surging the overall loss of the skeletal bone mass. In addition, CT is also
recognized as the steady inhibitor related to the osteoclast-mediated bone resorption
which makes it effective to be used in the therapeutic treatment related to
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 541
The CT activities are regulated with the use of calcitonin receptors which possess
high affinity. The calcitonin receptors are a member of G protein peptides of seven
transmembranes (Masi and Brandi 2007). All the members of the family have
identical structure along with seven other transmembranes spanning domain G
protein-coupled receptors. The cloning of porcine CTR (pCTR) cDNA was initially
done in the year 1991 by Lin et al. (1991). The characterization of the receptor is
based on its constituent of the extracellular long NH2-terminal domain. It shares
similarity with the parathyroid hormone which is linked with peptide receptor as well
as secretin receptor. Later, pCTR gene cloning showed that its length is about 70 kb
which also contains a minimum of 14 axons, in which 12 axons were inclusive of
protein. The cloning of the human CTR was done from ovarian cancer cell line, i.e.,
BIN-67. The different isoforms of CTR are the outcome of the gene splicing change
which has been indicated in various species of animals comprising divergent
transcripts of tissue expression as well as divergent signaling properties. Concerning
542 N. A. Nimer et al.
the large tumor cell of the bone, two kinds of isoforms have been explained, where
the first comprises similar design like GC-10, which is divergent from the earlier
explained human ovarian cancer CTR gene in the 50 region as it is devoid of the
71-bp segment while being almost similar to the 30 region. However, the second is
the CTR cDNA variant of the large human tumor cell, which is highlighted as GC-2,
devoid of the 71-bp 50 insert, though it comprises 48 encoded nucleotides part in the
first intercellular domain. The difference in the expression of CTR isoforms may be
the result of the biological regulatory mechanism which provides responses to the
calcitonin. The significant shift in the CTR isoforms can be explained as the
responsiveness of the variable related to calcitonin of the patients who have
increased turnover of the metabolic bone ailment. Moreover, the outlying of the
CTR gene is based on the chromosome 7q21.3.
With the several bone disorders, patients are being treated with calcitonin for the last
four decades, characterized on the basis of people that have high bone resorption
issues. The therapeutic function of calcitonin can be indicated through the patients
with disorders such as hypercalcemic states, i.e., bony metastases, Paget’s disease,
pain in osteoporotic fractures, toxicity of vitamin D, Sudeck’s atrophy, and osteopo-
rosis with high bone turnover. Until a few years ago, injection was the only possible
means of management. On the contrary, dosage forms are being produced, and a
calcitonin nasal spray is now commercially available using other routes
(Wimalawansa 2010). It seems difficult to administer calcitonin orally as it is a
peptide. However, in line with research studies into several delivery systems as well
as the increased improvisation of calcitonin molecule, it is highly probable to
administer the analogs of calcitonin orally. The process of oral administration can
be integrated through the buccal mucosal route. Bone pain that includes vertebral
fractures/osteolysis because of neoplasms is observed to be an indication for calci-
tonin therapy. Calcitonin seems to be successful for patients with Sudeck’s atrophy
and algoneurodystrophy. These are the syndromes caused by several factors such
as reflex dystrophy, post-traumatic osteoporosis, and iatrogenic neuropathy. More-
over, apart from the developed uses of calcitonin, the advantageous outcomes of
using calcitonin can also be observed, especially in the prevention of osteoporosis.
In case of osteoporosis that is associated with pregnancy, immobilization, and
increased bone catabolism, osteogenesis imperfecta, controlling loss of bone while
administering prednisone and heparin and severe renal insufficiency and excessive
osteoclastic activity is associated with these mentioned disorders. Calcitonin is
observed to be an additive therapy, although the growth of bone mass should be
increased when it comes to the ideal treatment for osteoporosis. Calcitonin and
bisphosphonates are important in controlling bone loss. In various osteoporosis
patients, albeit, that type of calcitonin serves as a less functional agent that is used
to provide stability to bone mass. In different types of osteoporosis, it seems to have
the greatest advantages when enhanced resorption is considered to be a feature. It has
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 543
15.9 Conclusion
expands to those fragments which modulate and at times counteract the parent
peptide response. Generally, the integration of the peptide fragments is frequent
for those receptors which are unidentified by the parent peptides. This chapter
explains the concepts related to the tachykinins, VIP-glucagon family,
neuropeptide Y, and calcitonin peptides which are present with the bioactive degra-
dation processes. The chapter demonstrates their related mechanisms of physiologi-
cal action, pathogenesis of the disease, pharmacology, receptors, and therapeutics.
The chapter provides insight that these neuropeptides can further be explored for
their utilization in drugs to improve the efficiency of the drugs as well as drug-like
development of the substance, which can serve as a rich source of development of
new pharmaceuticals.
References
Actelion (2011) Actelion and GSK discontinue clinical development of Almorexant [press release].
Available at: http://www.actelion.com/en/investors/media-releases/index.page?
newsId¼1483135. Accessed 3 Feb 2019
Amin K (2012) The role of mast cells in allergic inflammation. Respir Med 106(1):9–14. https://doi.
org/10.1016/j.rmed.2011.09.007. Epub 2011 Nov 22
Arcourt A, Gorham L, Dhandapani R, Prato V, Taberner FJ, Wende H, Gangadharan V,
Birchmeier C, Heppenstall PA, Lechner SG (2017) Touch receptor-derived sensory information
alleviates acute pain signaling and fine-tunes nociceptive reflex coordination. Neuron 93
(1):179–193
Atanasova KR, Reznikov LR (2018) Neuropeptides in asthma, chronic obstructive pulmonary
disease and cystic fibrosis. Respir Res 19(1):149. https://doi.org/10.1186/s12931-018-0846-4
Batista-Brito R, Vinck M, Ferguson KA, Chang JT, Laubender D, Lur G, Mossner JM, Hernandez
VG, Ramakrishnan C, Deisseroth K, Higley MJ, Cardin JA (2017) Developmental dysfunction
of VIP interneurons impairs cortical circuits. Neuron 95(4):884–895.e9
Belzung C, Yalcin I, Griebel G, Surget A, Leman S (2006) Neuropeptides in psychiatric diseases:
an overview with a particular focus on depression and anxiety disorders. CNS Neurol Disord
Drug Targets 5(2):135–145
Bik W, Wolinska-Witort E, Chmielowska M, Baranowska-Bik A, Rusiecka-Kuczalek E,
Baranowska B (2004) Vasoactive intestinal peptide can modulate immune and endocrine
responses during lipopolysaccharide-induced acute inflammation. Neuroimmunomodulation
11(6):358–364
Black PH (2002) Stress and the inflammatory response: a review of neurogenic inflammation. Brain
Behav Immun 16:622–653
Blanco EJ, Carretero-Hernández M, García-Barrado J, Iglesias-Osma MC, Carretero M, Herrero JJ,
Rubio M, Riesco JM, Carretero J (2013) The activity and proliferation of pituitary prolactin-
positive cells and pituitary VIP-positive cells are regulated by interleukin 6. Histol Histopathol
28(12):1595–1604
Bondy B, Baghai TC, Minov C, Schüle C, Schwarz MJ, Zwanzger P, Rupprecht R, Möller HJ
(2003) Substance P serum levels are increased in major depression: preliminary results. Biol
Psychiatry 53:538–542
Bourane S, Duan B, Koch SC, Dalet A, Britz O, Garcia-Campmany L, Kim E, Cheng L, Ghosh A,
Ma Q, Goulding M (2015) Gate control of mechanical itch by a subpopulation of spinal cord
interneurons. Science 350(6260):550–554
Campbell DE, Raftery N, Richard Tustin III, Tustin NB, DeSilvio ML, Cnaan A et al (2006)
Measurement of plasma-derived substance P: biological, methodological, and statistical
considerations. Clin Vaccine Immunol 13(11):1197–1203
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 545
Catalani E, De Palma C, Perrotta C, Cervia D (2017) Current evidence for a role of neuropeptides in
the regulation of autophagy. Biomed Res Int. https://doi.org/10.1155/2017/5856071
Chaiseha Y, Kang SW, Leclerc B, Kosonsiriluk S, Sartsoongnoen N, El Halawani ME (2010)
Serotonin receptor subtyopes influence prolactin secretion in the turkey. Gen Comp Endocrinol
165(1):170–175
Chandra R, Liddle RA (2015). Regulation of pancreatic secretion. Pancreapedia: Exocrine pancreas
knowledge base. https://doi.org/10.3998/panc.2015.38`
Chandrasekaran B (ed) (2013) Intelligence as adaptive behavior: an experiment in computational
neuroethology, vol 6. Academic Press, Cambridge
Chang MM, Leeman SE, Niall HD (1971) Amino-acid sequence of substance P. Nat New Biol 232
(29):86–87
Chapter MC, White CM, DeRidder A, Chadwick W, Martin B, Maudsley S (2010) Chemical
modification of class II G protein-coupled receptor ligands: frontiers in the development of
peptide analogs as neuroendocrine pharmacological therapies. Pharmacol Ther 125(1):39–54
Chen SR, Chen H, Zhou JJ, Pradhan G, Sun Y, Pan HL, Li DP (2017) Ghrelin receptors mediate
ghrelin-induced excitation of agouti-related protein/neuropeptide Y but not
pro-opiomelanocortin neurons. J Neurochem 142(4):512–520
Chi G, Huang Z, Li X, Zhang K, Li G (2018) Substance P regulation in epilepsy. Curr
Neuropharmacol 16(1):43–50
Chu HW, Kraft M, Krause JE, Rex MD, Martin RJ (2000) SP and its receptor neurokinin
1 expression in asthmatic airways. J Allergy Clin Immunol 106:713–722
Ciafrè S, Fiore M, Ceccanti M, Messina MP, Tirassa P, Carito V (2016) Role of neuropeptide
Tyrosine (NPY) in ethanol addiction. Biomed Rev 27:27–39
Coremans V, Ahmed T, Balschun D, D’Hooge R, DeVriese A, Cremer J, Cremer H (2010)
Impaired neurogenesis, learning and memory and low seizure threshold associated with loss
of neural precursor cell survivin. BMC Neurosci 11(1):2
Cox CD, Breslin MJ, Whitman DB, Schreier JD, McGaughey GB, Bogusky MJ, Roecker AJ,
Mercer SP, Bednar RA, Lemaire W, Bruno JG (2010) Discovery of the dual orexin receptor
antagonist [(7 R)-4-(5-chloro-1, 3-benzoxazol-2-yl)-7-methyl-1, 4-diazepan-1-yl] [5-methyl-2-
(2 H-1, 2, 3-triazol-2-yl) phenyl] methanone (MK-4305) for the treatment of insomnia. J Med
Chem 53(14):5320–5332
De Swert KO, Bracke KR, Demoor T, Brusselle GG, Joos GF (2009) Role of the tachykinin NK1
receptor in a murine model of cigarette smoke-induced pulmonary inflammation. Respir Res
10:37
Delgado M, Nieves V, Gonzalez-Rey E (2008) Vasoactive intestinal peptide protects against
β-amyloid-induced neurodegeneration by inhibiting microglia activation at multiple levels.
Glia 56(10):1091–1103
Diaz-delCastillo M, Woldbye DP, Heegaard AM (2018) Neuropeptide Y and its involvement in
chronic pain. Neuroscience 387:162–169
Dickson PV, Behrman SW (2013) Management of pancreatic neuroendocrine tumors. Surg Clin N
Am 93:675–691
Dickson L, Finlayson K (2009) VPAC and PAC receptors, “from ligands to function”. Pharmacol
Ther 121(3):294–316
Dickson L, Aramori I, McCulloch J, Sharkey J, Finlayson K (2006) A systematic comparison of
intracellular cyclic AMP and calcium signalling highlights complexities in human VPAC/PAC
receptor pharmacology. Neuropharmacology 51(6):1086–1098
Dorsam GP, Benton K, Failing J, Batra S (2011) Vasoactive intestinal peptide signaling axis in
human leukemi. World J Biol Chem 2(6):146–160
Eliska M, Peter K, Magdalena CD (2016) Role of substance P in the cardiovascular system.
Neuropeptides 58:41–51
Farzi A, Reichmann F, Holzer P (2015) The homeostatic role of neuropeptide Y in immune function
and its impact on mood and behaviour. Acta Physiol 213(3):603–627
546 N. A. Nimer et al.
Jiang W, Wang H, Li YS, Luo W (2016) Role of vasoactive intestinal peptide in osteoarthritis. J
Biomed Sci 23(1):63
Johansson E, Andersson L, Ornros J, Carlsson T, Ingeston-Carlsson C, Jansson S, Parillo L,
Zoppoli P, Barilla GO, Altschuler DL, Padula D, Lickert H, Fagman H, Nilsson M (2015)
Revising the embryonic origin of thyroid C cells in mice and humans. Development 142
(20):3519–3528
Kalina R, Gladkikh I, Dmitrenok P, Chernikov O, Koshelev S, Kvetkina A, Kozlov S,
Koslovskaya E, Monastyrnaya M (2018) New APETx-like peptided from sea anemone
Heteractus crispa modulate ASIC1a channels. Peptides 104:41–49
Kassim SK, ElTouny M, ElGuinaidy M, Abd ElMoghni MA, Abd ElMohsen A (2002) Serum
nitrates and vasoactive intestinal peptide in patients with gastroesophageal reflux disease. Clin
Biochem 35(8):641–646
Kastin A (2013) Handbook of biologically active peptides, 2nd edn. Academic Press, Cambridge,
pp 966–973
Koon HW, Zhao D, Na X, Moyer MP, Pothoulakis C (2004) Metalloproteinases and transforming
growth factor-alpha mediate substance P-induced mitogen-activated protein kinase activation
and proliferation in human colonocytes. J Biol Chem 279:45519–45527
Kramer MS, Cutler N, Feighner J, Shrivastava R, Carmen J, Sramek JS, Scott A (1998) Distinct
mechanism foir antidepressant activity by blockade of central substance P receptors. Science
281(5383):1640–1645
Laburthe M, Couvineau A, Tan V (2007) Class II G protein-coupled receptors for VIP and PACAP:
structure, models of activation and pharmacology. Peptides 28(9):1631–1639
Lai JP, Ho WZ, Zhan GX, Yi Y, Collman RG, Douglas S (2001) Substance P antagonist (CP-96,
345) inhibits HIV-1 replication in human mononuclear phagocytes. Proc Natl Acad Sci U S A
98:3970–3975
Lai JP et al (2008) Differences in the length of the carboxyl terminus mediate functional properties
of neurokinin-1 receptor. Proc Natl Acad Sci U S A 105:12605–12610. https://doi.org/10.1073/
pnas.0806632105
Larhammar D, Beck-Sickinger A, Colmers WF, Cox HM, Doods HN, Herzog H, Michel CM,
Quirion R, Schwartz T, Westfall T (2015) Neuropeptide Y receptors, introduction. Last
modified on 10/08/2015. Accessed 01 Feb 2019. IUPHAR/BPS Guide to PHARMACOLOGY,
http://www.guidetopharmacology.org/GRAC/FamilyIntroductionForward?familyId¼46
Lee FY, Lin HC, Tsai YT, Chang FY, Lu RH, Hou MC, Li CP, Chu CJ, Wang SS, Lee SD (1997)
Plasma SP levels in patients with liver cirrhosis: relationship to systemic and portal hemody-
namics. Am J Gastroenterol 92(11):2080–2084
Levinson DF, Duan J, Oh S, Wang K, Sanders AR, Shi J, Zhang N, Mowry BJ, Olincy A, Amin F
(2011) Copy number variants in schizophrenia: confirmation of five previous findings and new
evidence for 3q29 microdeletions and VIPR2 duplications. Am J Psychiatry 168:302–316
Lin H, Harris T, Flannery M, Aruffo A, Kaji E, Gorn A, Kolakowski L, Lodish H, Goldring SR
(1991) Expression cloning of an adenylate cyclase-coupled calcitonin receptor. Science 254
(5034):1022–1024
Litvak DA, Iseki H, Evers BM, Greely GH, Hellmich MR, Iwase K (1999) Characterization of two
novel proabsortive peptide YY analogs, BIM-43073 and BIM-43004C. Dig Dis Sci 44
(3):643–648
Liu J, Hu JH, Zhu QG, Li FQ, Wang J, Sun HJ (2007) Effect of matrine on the expression of
substance P receptor and inflammatory cytokines production in human skin keratinocytes and
fibroblasts. Int Immunopharmacol 7(6):816–823
Loh K, Herzog H, Shi YC (2015) Regulation of energy homeostasis by the NPY system. Trends
Endocrinol Metab 26(3):125–135
Lorente L, Martín MM, Pérez-Cejas A, Ferreres J, Solé-Violán J, Labarta L, Díaz C, Jiménez A
(2017) Sustained low serum substance P levels in non-surviving septic patients. Int J Mol Sci 18
(7):1531. https://doi.org/10.3390/ijms18071531
548 N. A. Nimer et al.
Lorente L, Rodriguez ST, Sanz P, Pérez-Cejas A, Padilla J, Díaz D et al (2018) Patients with high
serum substance P levels previously to liver transplantation for hepatocellular carcinoma have
higher risk of one-year liver transplantation mortality. Oncotarget 9(30):21552–21559
Lu CT, Jin RR, Jiang YN, Lin Q, Yu WZ, Mao KL, Tian FR, Zhao YP, Zhao YZ (2015) Gelatin
nanoparticle-mediated intranasal delivery of substance P protects against 6-hydroxydopamine-
induced apoptosis: an in vitro and in vivo study. Drug Des Devel Ther 9:1955–1962
Ludwig M (2011) Are neuropeptides brain hormones? J Neuroendocrinol 23(4):381–382
Majkowska-Pilip A, Halik PK, Gniazdowska E (2019) The significance of NK1 receptorligands and
their application in targeted radionuclide tumour therapy. Pharmaceutics 11(9):443
Manak MM, Moshkoff DA, Nguyen LT, Meshki J, Tebas P, Tuluc F, Douglas SD (2010) Anti-
HIV-1 activity of the neurokinin-1 receptor antagonist aprepitant and synergistic interactions
with other antiretrovirals. AIDS (London, England) 24(18):2789–2796
Marchi N, Granata T, Ghosh C, Janigro D (2012) Blood–brain barrier dysfunction and epilepsy:
pathophysiologic role and therapeutic approaches. Epilepsia 53(11):1877–1886
Masi L, Brandi ML (2007) Calcitonin and calcitonin receptors. Metabolism 4(2):117–122
Mathioudakis AG, Chatzimavridou-Grigoriadou V, Evangelopoulou E, Mathioudakis G (2013)
Vasoactive intestinal peptide inhaled agonists: potential role in respiratory therapeutics.
Hippokratia 17(1):12–16
Mawe GM, Hoffman JM (2013) Serotonin signalling in the gut – functions, dysfunctions and
therapeutic targets. Nat Rev Gastroenterol Hepatol 10(8):473–486
McQuaid KR (2018) Drugs used in the treatment of gastrointestinal diseases. In: Katzung BG
(ed) Basic and clinical pharmacology, 14th edn. McGraw Hill, New York, p 1105
Mendlewicz J, Oswald P, Claes S, Massat I, Souery D, Van Broeckhoven C, Del-Favero J (2005)
Patient-control association study of substance P-related genes in unipolar and bipolar affective
disorders. Int J Neuropsychopharmacol 8(4):505–513
Mittapalli GK, Roberts E (2014) Ligands of the neuropeptide Y Y2 receptor. Res Gate 24:430–441
Moreno D, Gourlet P, De Neef P, Cnudde J, Waelbroeck M, Robberecht P (2000) Development of
selective agonists and antagonists for the human vasoactive intestinal polypeptide VPAC
(2) receptor. Peptides 21(10):1543–1549
Muñoz M, Coveñas R (2013) Safety of neurokinin-1 receptor antagonists. Expert Opin Drug Saf 12
(5):673–685. https://doi.org/10.1517/14740338.2013.804059]
Nathoo AN, Moeller RA, Westlund BA, Hart AC (2001) Identification of neuropeptide-like protein
gene families in Caenorhabditis elegans and other species. PNAS 98(24):14000–14005
Neumann JM, Couvineau A, Murail S, Lacapere JL, Marc NJ (2008) Class-B GPCR activation: is
ligand helix-capping the key? Trends Biochem Sci 33(7):314–319
Niedermair T, Kuhn V, Doranehgard F, Stange R, Wieskötter B, Beckmann J et al (2014) Absence
of substance P and the sympathetic nervous system impact on bone structure and chondrocyte
differentiation in an adult model of endochondral ossification. Matrix Biol 38:22–35
Nilsson J, von Euler AM, Dalsgaard C-J (1985) Stimulation of connective tissue cell growth by
substance P and substance K. Nature 315(6014):61–63
Nussey SS, Whitehead SA (2013) Endocrinology: an integrated approach. CRC Press,
Philadelphia, PA
Opolka A, Straub RH, Pasoldt A, Grifka J, Grassel S (2012) Substance P and norepinephrine
modulate murine chondrocyte proliferation and apoptosis. Arthritis Rheum 64:729–739
Owen RT, Castaner R, Bolos J, Estivill C (2009) Almorexant dual orexin OX1/OX2 antagonist
treatment of sleep disorders. Drugs Future 34(1):5–10
Parent J, Kron M (2012) Jasper’s basic mechanisms of the epilepsies - neurogenesis and epilepsy,
4th edn. National Center for biotechnology information (US), Bethesda, MD
Pati S, Alexopoulos AV (2010) Pharmacoresistant epilepsy: from pathogenesis to current and
emerging therapies. Cleve Clin J Med 77(7):457–567
Pennefather JN, Lecci A, Candenas ML et al (2004) Tachykinins and tachykinin receptors: a
growing family. Life Sci 74:1445–1463
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 549
Smith SR, Weissman NJ, Anderson CM, Sanchez M, Chuang E, Stubbe S, Behavioral Modification
and Lorcaserin for Overweight and Obesity Management (BLOOM) Study Group (2010)
Multicenter, placebo-controlled trial of lorcaserin for weight management. N Eng J Med 363
(3):245–256
Spitsin S, Tebas P, Barrett JS, Pappa V, Kim D, Taylor D, Evans DL, Douglas SD (2017)
Antiinflammatory effects of aprepitant coadministration with cART regimen containing ritona-
vir in HIV-infected adults. JCI insight 2(19):e95893. https://doi.org/10.1172/jci.insight.95893.
Advance online publication
Steinhoff MS, von Mentzer B, Geppetti P, Pothoulakis C, Bunnett NW et al (2014) Tachykinins and
their receptors: contributions to physiological control and the mechanisms of disease. Physiol
Rev 94(1):265–301
Stojilkovic SS, Tabak J, Bertram R (2010) Ion channels and signaling in the pituitary gland. Endocr
Rev 31(6):845–915
Takei Y, Ando H, Tsutsui K (2016) Handbook of hormones: comparative endocrinology for basic
and clinical research. The Japan Society for Comparative Endocrinology; The University of
Tokyo, Chiba, Tokyon
Tang B, Yong X, Xie R, Li Q-W, Yang S-M (2014) Vasoactive intestinal peptide receptor-based
imaging and treatment of tumors. Int J Oncol 44(4):1023–1031
Taylor BK, Fu W, Kuphal KE, Stiller C-O, Winter MK, Corder GF, Urban JH, McCarson KE,
Marvizon JC (2014) Inflammation enhances Y1 receptor signaling, neuropeptide Y-mediated
inhibition of hyperglasia, and substance P release from primary afferent neurons. Neuroscience
256:178–194
Tekiner H, Acer N, Kelestimur F (2015) Sella turcica: an anatomical, endocrinological, and
historical perspective. Pituitary 18(4):575–578
Tian L, Cai L, Kang J (2000) Elevated substance P content in sputum and plasma in patients with
COPD and its relationship with FEV1/FVC. Zhonghua Jie He He Hu Xi Za Zhi 23:138–140
Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BK,
Hashimoto H, Galas L, Vaudry H (2009) Pituitary adenylate cyclase-activating polypeptide and
its receptors: 20 years after the discovery. Pharmacol Rev 61(3):283–357
Veldkamp MW, Geuzebroek GSC, Baartscheer A, Verkerk AO, Schumacher CA et al (2018a)
Neurokinin-3 receptor activation selectively prolongs atrial refractoriness by inhibition of a
background K+ channel. Nat Commun 9:4357. https://doi.org/10.1038/s41467-018-06530-5
Veldkamp MW, Geuzebroek G, Baartscheer A, Verkerk AO, Schumacher CA, Suarez GG, Berger
WR, Casini S, van Amersfoorth S, Scholman KT, Driessen A, Belterman C, van Ginneken A, de
Groot JR, de Bakker J, Remme CA, Boukens BJ, Coronel R (2018b) Neurokinin-3 receptor
activation selectively prolongs atrial refractoriness by inhibition of a background K+ channel.
Nat Commun 9(1):4357
Verma AK, Manohar M, Upparahalli Venkateshaiah S, Mishra A (2017) Neuroendocrine cells
derived chemokine vasoactive intestinal polypeptide (VIP) in allergic diseases. Cytokine
Growth Factor Rev 38:37–48
Vezzani A, French J, Bartfai T, Baram T (2011) The role of inflammation in epilepsy. Nat Rev
Neurol 7:31–40
Vezzani A, Friedman A, Dingledine RJ (2013) The role of inflammation in epileptogenesis.
Neuropharmacology 69:16–24
Vilisaar J, Arsenescu RI (2016) Roles of substance P in gastrointestinal functions and neuroimmune
interactions. In: Constantinescu C, Arsenescu R, Arsenescu V (eds) Neuro-immuno-gastroen-
terology. Springer, Cham, pp 53–73
von Zastrow M (2018) Drug receptors & pharmacodynamics. In: Katzung BG (ed) Basic and
clinical pharmacology, 14th edn. McGraw Hill, New York, pp 20–41. Gray
Vu J, Larauche M, Fores M, Luong L, Norris J, Oh S, Li-Jung L, Waschek J, Pisegna J, Germano P
(2015) Regulation of apetite, biody composition and metabolic hormones by vasoactive intesti-
nal polypeptide (VIP). J Mol Neurosci 56(2):377–387
15 Pharmacology of Neuropeptides: Substance P, Vasoactive Intestinal Peptides,. . . 551
Wang L, Singh M, Bonewald LF, Detamore MS (2009) Signalling strategies for osteogenic
differentiation of human umbilical cord mesenchymal stromal cells for 3D bone tissue engi-
neering. J Tissue Eng Regen Med 3:398–404
Wang XF, Ge TT, Fan J, Yang W, Cui RJ (2017) The role of substance P in epilepsy and seizure
disorders. Oncotarget. 8(44):78225–78233
White CM, Ji S, Cai H, Maudsley S, Martin B (2010) Therapeutic potential of vasoactive intestinal
peptide and its receptors in neurological disorders. CNS Neurol Disord Drug Targets 9
(5):661–666
Wimalawansa SJ (2010) Calcitonin: history, physiology, pathophysiology and therapeutic
applications. https://doi.org/10.1016/B978-0-12-374602-300053-5
Wu D, Lee D, Sung YK (2011) Prospect of vasoactive intestinal peptide therapy for COPD/PAH
and asthma: a review. Respir Res 12(1):45
Xiao J, Yu W, Wang X, Wang B, Chen J, Liu Y, Li Z (2016) Correlation between neuropeptide
distribution, cancellous bone microstructure and joint pain in postmenopausal women with
osteoarthritis and osteoporosis. Neuropeptides 56:97–104
Yin J, Chapman K, Clark LD, Shao Z, Borek D, Xu Q et al (2018) Rosenbaum crystal structure of
the human NK1 tachykinin receptor. Proc Natl Acad Sci U S A 115(52):13264–13269
Zalecki M (2019) Gastric ulcer induced changes in substance P and Nk1, Nk2, Nk3 receptors
expression in different stomach localizations with regard to intrinsic neuronal system.
Histochem Cell Biol 151(1):29–42
Zikou A, Sioka C, Alexiou GA, Fotopoulos A, Voulgaris S, Argyropoulou MI (2018) Radiation
Necrosis, pseudoprogression, pseudoresponse, and tumor recurrence: imaging challenges for
the evaluation of treated gliomas. Contrast Media Mol Imaging 2018:1–6
Neuropeptides and Neurotransmission
16
Anindita Mondal Gantait, Yazan A. Bataineh, Hiba Salim Surchi,
Arunava Gantait, G. Tulja Rani, Paramita Paul, Sarah Falah Kokaz,
Bilal A. Al-Jaidi, Puneet Kumar, Saumen Karan,
and Tanushree Singha
Abstract
Over the past four decades, many neuropeptides, that is, 3–100 amino-acid-long
polypeptides, have been identified in the central nervous system and the periph-
eral nervous system which can act on either neural substrates such as neurons and
glial cells or other target cells. Neuropeptides mediate neuronal communication
by acting on neuropeptide receptors. Neuropeptide receptors include over
44 receptor families, of which most are G protein-coupled receptors.
Neuropeptides and their cognate receptors are involved in various physiological
and pathophysiological functions, such as pain regulation, blood pressure, body
Keywords
Neuropeptide · VIP · Substance P · NPY · Endorphin · Enkephalin · Dynorphin
Abbreviations
5-HT Serotonin
ACE Angiotensin-converting enzyme
Ach Acetylcholine
ACTH Adrenocorticotrophic hormone
a-MSH Melanocyte-stimulating hormone
CAMP Cyclic Adenosine Monophosphat
CGRPs Calcitonin gene-related peptides
CINV Chemotherapy induced nausea and vomiting
CNS Central Nervous System
DAG Diacylglycerol
DOR δ opioid receptor
EPI Epinephrine
GABA Gamma amino butyric acid
GIP GPCR-interacting proteins
GLP-1 Glucagon-like peptide-1
GPCR G protein-coupled receptors
HA Histamine
IBD Inflammatory bowel disease
IP3 Inositol triphosphate
KOR κ opioid receptor
MAPK Mitogen-activated protein kinase
MEC Moderately emetogenic chemotherapy
MOR μ-opioid receptors
NE Noradrenaline
NK Neurokinin
NKA Neurokinin A
16 Neuropeptides and Neurotransmission 555
NMDA N-methyl-D-aspartate
NPY Neuropeptide Y
PACAP Pituitary adenylate cyclase-activating polypeptide
PC2 Pro-protein convertase-2
PENK-A Proenkephalin-A
PHM Peptide histidine methionine
POMC Precursor proopiomelanocortin
PP Pancreatic polypeptide
PPT-A PreprotachykininA
PYY Peptide YY
RAMPs Receptor activity modifying proteins
SP Substance P
VIP Vasoactive intestinal peptides
16.1 Introduction
Neurotransmitters are endogenous substances that are released from the neuron.
These molecules are responsible for the transmission of information on chemical
synapses. Neurotransmitters are released from the presynaptic axonal membrane to
the synaptic cleft and bind to the receptors present in the postsynaptic membrane of
other neurons and conduct signal transfer across the synapse. A molecule can be
considered a neurotransmitter if it fulfills the following criteria. It must present in the
vesicles along with the suitable enzymes that help in their synthesis and breakdown.
The substance should be released from the nerve ending in a chemically or
pharmacologically identifiable form as an effect of presynaptic nerve stimulation.
They must have action in the synapse and its actions must be prevented by receptor
blockers. Thus the neurotransmitters are chemicals that are released from the neuron
on nerve stimulation by generation of action potential and communicate with the
muscle, other organs and other neurons (Purves et al. 2001; González-Espinosa and
Guzmán-Mejía 2014; Fieber 2017).
The process of neurotransmission takes place in the synaptic cleft of the nervous
system composed of the basic cells called neurons. Neurons, in spite of having
unimaginable variation in their structure and functions, possess many common
features among them, and they do the work of cellular communication (Patri 2019;
Lodish et al. 2000). The communication between two neurons starts when electrical
impulse or action potential is carried in only one direction by the long slender
projection called the axon (Hormuzdi et al. 2004). The action potential reaches the
axon terminal but cannot cross the synaptic space and thus triggers the neurotrans-
mitter release from their storage vesicles present in synaptic terminals (presynaptic
membrane) into a space known as the synapse. The neurotransmitters then bind with
special proteins called the receptors present in the postsynaptic membrane of another
neuron, which triggers the action potential to move toward the cell body to the axon
of the postsynaptic neuron (Lodish et al. 2000). The neurotransmitters then release a
related message from the receptor into the synaptic space. Some of the
neurotransmitters degraded by the local enzymes are taken back by the transporter
proteins present in the presynaptic membrane (Lodish et al. 2000; Forehand 2009).
The neurotransmitters that are taken back are repackaged into a vesicle which is
released again when an action potential reaches the axon terminal. The entire process
is repeated when an action potential reaches the axon terminal (Forehand 2009).
16.3 Substance P
extracted from equine horse brain and gut tissues (V Euler and Gaddum 1931). This
substance was later named Substance P, P as in the powder obtained during the
procedure. Substance P is an undecapeptide, composed of a chain of 11 amino acid
residues (Arg-Pro-Lys-Gln-Gln-Phe-Phe-Gly-Leu-Met-NH2). It shares the same
carboxyl terminal sequence, Phe-(Phe or Val)-Gly-Leu-Met-NH2, with the rest of
the tachykinin family members; however, it has a net positive charge. The
N-terminus holds the positively charged residues, while the C-terminus is confined
to hydrophobic residues, rendering it an amphiphilic peptide. The biological actions
are mediated by neurokinin (NK) G protein-coupled receptors acting on smooth
muscle contraction, vasodilation, nociception and modulation of inflammatory
responses (Mashaghi et al. 2016a, b).
16.3.1 Physiology
16.3.2 Pathophysiology
Due to its wide distribution in the body, substance P has numerous physiological
implications in the CNS, cardiovascular, respiratory, gastrointestinal systems and
modulation of the immune response. Additionally, it plays a critical role in the
regulation of the excitability of dorsal horn nociceptive neurons (Christofi 2018). It
is associated with the regulation of mood disorders, anxiety, stress, neurotoxicity and
pain. In the gastrointestinal tract, SP and other tachykinins act as neurotransmitters
that regulate motor activity, secretion of ions and vascular functions. Therefore, the
elevation of the neuropeptide or its receptor has been linked to several chronic
pathological conditions such as inflammatory disorders (e.g. irritable bowel syn-
drome) (Sohn et al. 2013), Alzheimer’s disease (Severini et al. 2016), Parkinson’s
disease (Thornton and Vink 2015), and mood and mental disorders. Nerve fibers
producing substance P were detected both in atrial and ventricular myocardia, where
efferent and afferent sensory functions of the peripheral neurons innervating the
heart and coronary arteries are mediated by tachkykinins and calcitonin gene-related
peptides (CGRPs). It has been pointed out that substance P possesses binding sites
surrounding coronary arteries and cardiac fibers and on mitral and tricuspid valves. It
has negative inotropic and chronotropic effects due to an increase in the activity of
cholinergic neurons, leading to a weaker heart rate and a change in the force of
contractions (Mistrova et al. 2016). SP and its receptors’ distribution in the brain was
first described in the 1970s. It has been demonstrated that high levels of SP
immunoreactivity have been spotted in areas associated with regulation of stress
and anxiety reactions, mainly in the hippocampus, amygdala and some areas of the
hypothalamus. In these aforementioned regions, the peptide coexists in the same
neuron terminal with various neurotransmitters and neuropeptides such as glutamate,
GABA, histamine, acetylcholine, dopamine and serotonin (Ebner and Singewald
2006).
Substance P initiates the expression of various known immunological chemical
messengers. Primary lymphoid organs such as the bone marrow and thymus and
secondary ones such as the spleen and lymph nodes have been observed to be
innervated by neurons containing SP. This suggests that substance P may act as a
16 Neuropeptides and Neurotransmission 559
Chemotherapy
Medulla oblongata
Increased afferent input to the chemoreceptor trigger zone and vomiting center
16.3.4.1 Aprepitant/Fosaprepitant
16.3.4.2 Pharmacokinetics
F F
F F
O F
O O F
N N F
O
HN O O N
F N HO
H N
P
F F N
HO F
F
F NH
F
O
Aprepitant Fosaprepitant
is coated and encapsulated. A dose of 125 mg of aprepitant is given on the first day,
followed by 80 mg once daily. Onset of action is 1 h when administered orally, with
peak plasma level at 4 h. Aprepitant has a bioavailability of 60–70% after oral
administration, indicating a low liver first pass extraction.
16.3.5 Rolapitant
NH
O
F
F
H F
Rolapitant
16.3.5.2 Pharmacokinetics
Vasoactive intestinal peptide (VIP) is the most abundant neuropeptide in the gut,
controlling intestinal motility and water and electrolyte secretion. VIP belongs to the
glucagon/secretin superfamily; it was isolated from porcine small intestine and
shown to have a similar amino acid sequence than glucagon and secretin (Said and
Mutt 1970; Mutt and Said 1974). VIP not only functions as a peptide neurotransmit-
ter in the gastrointestinal tract but also in the central and peripheral nervous systems.
16 Neuropeptides and Neurotransmission 563
16.4.1 Physiology
C122 K127
N28
N24
NH2 G116
Y22
N-ted
Q135
D107 F6
F0
Fig. 16.4 3D structural model of VPAC1 receptor showing different binding pocket for VIP.
(Reprinted with permission from Couvineau et al. 2013)
564 A. M. Gantait et al.
16.4.2 Pathophysiology
16.4.3 Pharmacology
GPCRs. These class B or class II receptors for peptides bear low sequence
homologies with other members of the superfamily of GPCRs. VPAC receptors
show several common properties with other class II GPCRs such as a large
N-terminal extracellular domain (>120 residues) with 10 highly conserved amino
acids including six cysteines and several potential N-glycosylation sites. Secretin,
helodermin, GRF and PHM (peptide histidine methionineamide), the other members
of the VIP-secretin structural family, can also bind to VPAC1 or VPAC2 receptors
with less affinity (Laburthe et al. 2002). Agonists and antagonists of VPAC1 and
VPAC2 receptors are described in Table 16.1.
16.5 Neuropeptide Y
16.5.1 Physiology
are mediated by at least five types of receptors that have been identified, Y1, Y2, Y4,
Y5 and Y6; however, Y6 has not been assigned to any biological function. Addi-
tional distinguished receptor subtypes exist in other species. The Y receptors belong
to the G protein-coupled receptor family, exhibiting its effects through inhibiting
cAMP-dependent kinase. NPY plays vital roles in the gut-brain axis
communications, by being synthesized in the brain and acting locally in a paracrine
fashion in the GIT through its receptors to regulate motility and electrolyte secretion.
It thus regulates appetite and metabolism (Cox 2007).
16.5.2 Pathophysiology
The local action of NPY-like peptides after their exocytotic release depends on their
concentration, receptor selectivity, the expression of Y receptors and the presence of
specific peptidases influencing half-life (von Hörsten et al. 2004). The potential roles
of NPY in the etiology and pathophysiology of mood and anxiety disorders, as well
as related alcohol use disorders, have been extensively studied. Thus, modulation of
NPY-ergic activity within the CNS, via ligands aimed at different receptor subtypes,
may be an attractive target for treatment development for affective disorders, as well
as for alcohol use disorders (Thorsell and Mathé 2017). NPY has been found to
stimulate intake of food, preferably carbohydrate intake. It decreases latency to eat,
increases the drive to eat and delays satiety by augmenting meal size (Beck 2006).
The interactions between the immune system, that is, immune cells and gut
neurohormones, especially NPY, play an important role in producing inflammation
in inflammatory bowel disease (IBD). Thus, NPY may be targeted to diminish
the inflammation in IBD (El-Salhy and Hausken 2016). The significance of
investigating the underlying pathophysiological mechanisms of arteriosclerotic car-
diovascular disease and the role of NPY in this regard is great. It is involved in the
pathogenesis of arteriosclerosis via aggravating endothelial dysfunction and growth
of vascular smooth muscle cells, by the formation of foam cells and platelet
aggregation (Zhu et al. 2016; Sun et al. 2017). NPY, a major peripheral vascular
contractive neurotransmitter, interacts with its receptors and is thus associated
with the pathology and development of diabetes. It further contributes to diabetes-
induced cardiovascular disease by promoting the proliferation of endothelial cells
and vascular fibrosis (Sun et al. 2017). The effects of NPY on different species are
described in Table 16.2.
16.5.3 Pharmacology
NPY is one of the most effective orexigenic peptides that are mostly found in the
brain (Beck 2006; Hofmann et al. 2019). NPY acts via four different types of
functional G protein-coupled receptors (GPCRs) known as Y1, Y2, Y4 and Y5
(Table 16.3). All of these NPY receptors act via coupling to Gi and thus by inhibiting
the synthesis of cAMP. Y1 is mainly distributed to the brain, that is, anterior
16 Neuropeptides and Neurotransmission 567
The endogenous opioids system includes numerous peptides that are widely
distributed throughout the human body, all acting as ligands for opioid receptors
including endorphins, enkephalins, dynorphins and, most recently discovered,
endomorphins, all of which are biologically active products that are released at the
synaptic terminals of opioidergic neurons. Classical opioid receptors are divided into
three subtypes, the μ receptor (MOR), δ receptor (DOR) and κ receptor (KOR), all
belonging to the GPCR family, and consisting of highly homologous
7-transmembranous helices linked with extracellular peptide loops (Li et al. 2012).
Opioid neurotransmission appears to affect many CNS functions, such as
nociception, cardiovascular regulation, respiratory rate, neuroendocrine activity,
aggressive, locomotive, pleasure and sexual behaviours, and learning and memory.
16.6.2 Endorphins
Endorphins are natural endogenous opioid neuropeptides, are one of the major
products of the precursor proopiomelanocortin (POMC) and are secreted by the
anterior pituitary gland through the hypothalamus in response to certain physiologi-
cal triggers such as strenuous physical exercise, stress and pain, and they resemble
opiates in their ability to produce analgesic effects and inhibit transmission of pain
signals. Other active products of this precursor include adrenocorticotrophic hor-
mone (ACTH) and a-melanocyte-stimulating hormone (a-MSH). The word endor-
phin is contracted from the words Endogenous and morphine. Four types of
endorphins are produced in the human body, α-, β-, γ- and σ- endorphins, each
having different numbers and types of amino acids in their molecules, between
16 and 31 amino acid residues in each peptide molecule (Shrihari 2017; Li et al.
2012).
Endorphins are widely distributed in many parts of the body, mainly in the
pituitary glands as well as in the brain. β-endorphin is the most potent and abundant,
in terms of natural pain relief, and is present in the neurons of both the central and
peripheral nervous system. They are released during pain or stress, and are
associated with sexual and maternal behavior. Additionally, endorphins have been
found to be associated with states of pleasure including emotions brought about by
laughter, intercourse, love and even appetizing food (Sprouse-Blum et al. 2010).
16 Neuropeptides and Neurotransmission 569
in cases of opioid overdose to mitigate bodily response to the opioid (Glanz et al.
2018; Gonzalez and Brogden 1988). Another example is alvimopan, as it has a high
affinity for MOR, but low systemic absorption. High concentrations of opioid
receptors can be found throughout the gastrointestinal tract, and stimulation of
these receptors by opioid analgesics has a direct local effect on bowel function and
motility (Vaughan-Shaw et al. 2012). Reports have shown that alvimopan accelerates
the gastrointestinal recovery period and therefore it is indicated for patients
undergoing radical cystectomy for bladder cancer, which is associated with delayed
gastrointestinal recovery that prolongs hospital stay (Cheryl T. Lee et al. 2014).
Other MOR antagonists include levallorphan and nalmefene, the latter being clini-
cally used to reduce alcohol dependence (Clément Palpacuer et al. 2015). In com-
parison to chronically morphine-treated mice, repeated peptidomimetic-treated mice
developed less analgesic tolerance and/or physical dependence. Analysis of in vitro
and in vivo data has permitted structure activity relationships to guide further
discovery and chemical synthesis of opioid analgesics that suggest improved
clinical use.
16.6.3 Enkephalins
Enkephalins are pentapeptides first discovered and isolated from porcine brain
tissues in 1975, involved in the regulation of pain. There are two forms of
enkephalins, Leu-enkephalin (Tyr-Gly-Gly-Phe-Leu) containing the amino acid
leucine, and Met-enkephalin (Tyr-Gly-Gly-Phe-Met) containing methionine
(Comb et al. 1982), both generated from a common precursor proenkephalin-A
(PENK-A) by proteolytic enzymes. Leu-enkephalin and met-enkephalin are
metabolized by several enzymes called enkephalinases that include endopeptidases,
aminopeptidases and angiotensin-converting enzyme (Thanawala et al. 2008),
among others. Both enkephalin forms exert their effects through the δ-opioid
receptor (DOR) that belongs to the GPCR family. Met-enkephalin is found mainly
in the adrenal medulla and the brain acting as a neurotransmitter/neuromodulator and
as an active regulator of cell proliferation. Like μ-receptors, δ-opioid receptor
signaling investigations have primarily focused on mechanisms of opioid analgesia
(Al-Hasani and Bruchas 2011). Despite MOR-based analgesics being potent and
efficient in alleviating acute severe pain, they are ineffective in treating chronic pain
syndromes, which is why the attention has been driven to other receptors,
particularily δ-receptors, showing to be potential targets for developing novel opiate
analgesics for chronic pain management. Nonetheless, initiating tolerance is a
limitation for their use as long-acting opioid analgesics (Charfi et al. 2015).
DPDPE is one of the early synthetic DOR agonists developed, and is structurally
similar to met-enkephalin (Bilsky et al. 1995). SNC80 is the first non-peptidic-
selective DOR ligand developed and it successfully produced antidepressant, anxio-
lytic and analgesic effects, but its use was limited due to causing convulsions when
administered in high doses (Dripps et al. 2018). Other agonists include BMS986187,
a potent δ-opioid receptor-positive allosteric modulator that enhances the affinity of
leu-enkephalin to the δ-opioid receptor, yet it has a low potency due to limited
16 Neuropeptides and Neurotransmission 571
16.6.4 Dynorphins
16.7 Conclusion
This chapter describes some of the different classes of neuropeptides. The biosyn-
thesis and metabolism with their physiology, pathophysiology and pharmacology
have been described. Several agonists and antagonists of these peptide receptors
continue to show great success in the development of therapeutic agents for the
treatment of a variety of disorders, which include cardiovascular, epilepsy, immune,
psychiatric, substance abuse and body weight disorder.
References
Aapro M, Hesketh PJ, Jordan K, Gralla RJ, Rossi G, Rizzi G, Palmas M (2016) Safety of an oral
fixed combination of netupitant and palonosetron (NEPA): pooled data from the phase II/III
clinical program. Oncologist 21(4):494–502
Al-Hasani R, Bruchas MR (2011) Molecular mechanisms of opioid receptor-dependent signaling
and behavior. Anesthesiology 115(6):1363–1381
Anderson RI, Lopez MF, Griffin WC, Haun HL, Bloodgood DW, Pati D, Boyt KM, Kash TL,
Becker HC (2019) Dynorphin-kappa opioid receptor activity in the central amygdala modulates
binge-like alcohol drinking in mice. Neuropsychopharmacology 44(6):1084–1092
Baraniuk JN, Silver PB, Kaliner MA, Barnes PJ (1992) Neuropeptide Y is a vasoconstrictor in
human nasal mucosa. J Appl Physiol 73(5):1867–1872
Beck B (2006) Neuropeptide Y in normal eating and in genetic and dietary-induced obesity. Philos
Trans R Soc Biol Sci 361(1471):1159–1185
Bilsky EJ, Calderon SN, Wang T, Bernstein RN, Davis P, Hruby VJ, McNutt RW, Rothman RB,
Rice KC, Porreca F (1995) SNC 80, a selective, nonpeptidic and systemically active opioid delta
agonist. J Pharmacol Exp Ther 273(1):359–366
Bockaert J, Roussignol G, Becamel C, Gavarini S, Joubert L, Dumuis A, Fagni L, Marin P (2004)
GPCR-interacting proteins (GIPs): nature and functions. Biochem Soc Trans 32(5):851–855
Broom DC, Jutkiewicz EM, Folk JE, Traynor JR, Rice KC, Woods JH (2002) Nonpeptidic δ-opioid
receptor agonists reduce immobility in the forced swim assay in rats.
Neuropsychopharmacology 26(6):744–755
Bruchas MR, Chavkin C (2010) Kinase cascades and ligand-directed signaling at the kappa opioid
receptor. Psychopharmacology 210(2):137–147
Bruno JB, Matos MH, Chaves RN, Figueiredo JR (2011) Involvement of vasoactive intestinal
peptide (VIP) of ovarian physiology. Anim Reprod 8(3/4):51–57
Burbach JP (2011) What are neuropeptides? Methods Mol Biol 789:1–36
Catalani E, De Palma C, Perrotta C, Cervia D (2017) Current evidence for a role of neuropeptides in
the regulation of autophagy. Biomed Res Int 2017:1–10
Charfi I, Audet N, BagheriTudashki H, Pineyro G (2015) Identifying ligand-specific signalling
within biased responses: focus on δ opioid receptor ligands. Br J Pharmacol 172(2):435–448
Chavkin C (2013) Dynorphin–still an extraordinarily potent opioid peptide. Mol Pharmacol 83
(4):729–736
Christofi FL (2018) TRPV1 sensory neurons and enteric glia in ENS link tachykinins to
Neuroinflammation and nociception. Cell Mol Gastroenterol Hepatol 6(3):354–355
Comb M, Seeburg PH, Adelman J, Eiden L, Herbert E (1982 Feb) Primary structure of the human
met-and Leu-enkephalin precursor and its mRNA. Nature 295(5851):663–666
Couvineau A, Tan Y-V, Ceraudo E, Laburthe M (2013) Strategies for studying the ligand binding
site of GPCRs. Methods Enzymol 520:219–237
Cox HM (2007) Peptide YY: a neuroendocrine neighbor of note. Peptides 28(2):345–351
16 Neuropeptides and Neurotransmission 573
Dalayeun JF, Nores JM, Bergal S (1993) Physiology of β-endorphins. A close-up view and a review
of the literature. Biomed Pharmacother 47(8):311–320
Dehlin HM, Levick SP (2014) Substance P in heart failure: the good and the bad. Int J Cardiol 170
(3):270–277
Delgado M, Pozo D, Ganea D (2004) The significance of vasoactive intestinal peptide in
immunomodulation. Pharmacol Rev 56(2):249–290
Drake CT, Chavkin C, Milner TA (2007) Opioid systems in the dentate gyrus. Prog Brain Res
163:245–814
Dripps IJ, Boyer BT, Neubig RR, Rice KC, Traynor JR, Jutkiewicz EM (2018) Role of signalling
molecules in behaviours mediated by the δ opioid receptor agonist SNC80. Br J Pharmacol. 175
(6):891–901
Ebner K, Singewald N (2006) The role of substance P in stress and anxiety responses. Amino Acids
31(3):251–272
El-Salhy M, Hausken T (2016) The role of the neuropeptide Y (NPY) family in the pathophysiology
of inflammatory bowel disease (IBD). Neuropeptides 55:137–144
Fan X, Markram H (2019) A brief history of simulation neuroscience. Front Neuroinform 13:32
Fieber LA (2017) Neurotransmitters and neuropeptides of invertebrates. In: The Oxford handbook
of invertebrate neurobiology. Oxford University Press, Oxford
Forehand CJ (2009) The action potential, synaptic transmission, and maintenance of nerve function.
In: Rhoades RA, Bell DR (eds) Medical physiology: principles for clinical medicine. Lippincott
Williams & Wilkins, A Wolters Kluwer Business, Philadelphia, pp 38–64
Fujiwara H, Kurihara N, Hirata K, Ohta K, Kanazawa H, Takeda T (1993) Effect of neuropeptide Y
on human bronchus and its modulation of neutral endopeptidase. J Allergy Clin Immunol 92
(1):89–94
Furness JB, Young HM, Pompolo S, Bornstein JC, Kunze WA, McConalogue K (1995)
Plurichemical transmission and chemical coding of neurons in the digestive tract. Gastroenter-
ology 108(2):554–563
Gehlert, D. R. (2009). Neuropeptide Y (NP Y) and its receptors. In: Encyclopedia of neuroscience.
Academic Press, Cambridge. pp. 837–842
Glanz JM, Narwaney KJ, Mueller SR, Gardner EM, Calcaterra SL, Xu S, Breslin K, Binswanger IA
(2018) Prediction model for two-year risk of opioid overdose among patients prescribed chronic
opioid therapy. J Gen Intern Med 33(10):1646–1653
Glass SM, Leddy SM, Orwin MC, Miller GP, Furge KA, Furge LL (2019) Rolapitant is a reversible
inhibitor of CYP2D6. Drug Metab Dispos 47(6):567–573
Gomariz RP, Juarranz Y, ABAD C, Arranz A, Leceta J, Martinez C (2006) VIP–PACAP system in
immunity: new insights for multitarget therapy. Ann N Y Acad Sci 1070(1):51–74
Gonzalez JP, Brogden RN (1988) NaltrexoneDrugs 35(3):192–213
González-Espinosa C, Guzmán-Mejía F (2014) Basic elements of signal transduction pathways
involved in chemical neurotransmission. In: Identification of neural markers accompanying
memory. Elsevier, Amsterdam, pp 121–133
Gourlet P, De Neef P, Cnudde J, Waelbroeck M, Robberecht P (1997) In vitro properties of a high
affinity selective antagonist of the VIP1 receptor. Peptides 18(10):1555–1560
Gozes I, Fridkin M, Hill JM, Brenneman DE (1999) Pharmaceutical VIP: prospects and problems.
Curr Med Chem 6:1019–1034
Granier S, Manglik A, Kruse AC, Kobilka TS, Thian FS, Weis WI, Kobilka BK (2012) Structure of
the δ-opioid receptor bound to naltrindole. Nature 485(7398):400–404
Grunberg S, Chua D, Maru A, Dinis J, DeVandry S, Boice JA, Hardwick JS, Beckford E, Taylor A,
Carides A, Roila F (2011) Single-dose fosaprepitant for the prevention of chemotherapy-
induced nausea and vomiting associated with cisplatin therapy: randomized, double-blind
study protocol—EASE. J Clin Oncol 29(11):1495–1501
Harmar AJ, Arimura A, Gozes I, Journot L, Laburthe M, Pisegna JR, Rawlings SR, Robberecht P,
Said SI, Sreedharan SP, Wank SA (1998) Nomenclature of receptors for vasoactive intestinal
peptide (VIP) and pituitary adenylate cyclase activating polypeptide (PACAP). Pharmacol Rev
50:265–270
574 A. M. Gantait et al.
Palpacuer C, Laviolle B, Boussageon R, Reymann JM, Bellissant E, Naudet F (2015) Risks and
benefits of nalmefene in the treatment of adult alcohol dependence: a systematic literature
review and meta-analysis of published and unpublished double-blind randomized controlled
trials. PLoS Med 12(12):e1001924
Patri M (2019) Synaptic transmission and amino acid neurotransmitters. In: Neurochemical basis of
brain function and dysfunction. IntechOpen, London
Purves D, Augustine GJ, Fitzpatrick D, Katz LC, LaMantia A-S, McNamara JO (eds) (2001)
Neuroscience, 2nd edn. Sinauer Associates, Sunderland, MA
Rapoport B, Schwartzberg L, Chasen M, Powers D, Arora S, Navari R, Schnadig I (2016) Efficacy
and safety of rolapitant for prevention of chemotherapy-induced nausea and vomiting over
multiple cycles of moderately or highly emetogenic chemotherapy. Eur J Cancer 57:23–30
Roos C, Dahlgren D, Berg S, Westergren J, Abrahamsson B, Tannergren C, Sjögren E, Lennernäs
H (2017 Aug 10) In vivo mechanisms of intestinal drug absorption from
aprepitantnanoformulations. Mol Pharm 14(12):4233–4242
Russo AF (2017) Overview of neuropeptides: a wakening the senses? Headache 57:37–46
Said SI (1967) Vasoactive substances in the lung. In: Proceedings of tenth Aspen emphysema
conference, Aspen, CO, June 7–10, 1967. U.S. Public Health Service Publication, vol. 1787,
pp. 223–228
Said SI (1989) Vasoactive intestinal polypeptide and asthma (editorial). New Engl J Med
320:1271–1273
Said SI, Mutt V (1970) Potent peripheral and splanchnic vasodilator peptide from normal gut.
Nature 225(5235):863–864
Said SI, Rattan S (2004) The multiple mediators of neurogenic smooth muscle relaxation. Trends
Endocrinol Metab 15:189–191
Said SI, Berisha HI, Pakbaz H (1996) Excitotoxicity in lung: N-methyl-D-aspartate-induced, nitric
oxide-dependent, pulmonary edema is attenuated by vasoactive intestinal peptide and by
inhibitors of poly (ADP-ribose) polymerase. Proc Natl Acad Sci U S A 93:4688–4692
Said SI, Hamidi SA, Dickman KG, Szema AM, Lyubsky S, Lin RZ, Jiang YP, Chen JJ, Waschek
JA, Kort S (2007) Moderate pulmonary arterial hypertension in male mice lacking the vasoac-
tive intestinal peptide gene. Circulation 115(10):1260–1268
Seidah NG, Day R, Marcinkiewicz M, Chretien M (1998) Precursor convertases: an evolutionary
ancient, cell-specific, combinatorial mechanism yielding diverse bioactive peptides and
proteins. Ann N Y Acad Sci 839(1):9–24
Severini C, Petrella C, Calissano P (2016) Substance P and Alzheimer’s disease: emerging novel
roles. Curr Alzheimer Res 13(9):964–972
Sherwood NM, Krueckl SL, McRory JE (2000) The origin and function of the pituitary adenylate
cyclase-activating polypeptide (PACAP)/glucagon superfamily. Endocr Rev 21(6):619–670
Shrihari TG (2017) Endorphins on cancer: a novel therapeutic approach. J Carcinog Mutagen 8:298
Sohn W, Lee OY, Lee SP, Lee KN, Jun DW, Lee HL, Yoon BC, Choi HS, Sim J, Jang K-S (2013)
Mast cell number, substance P and vasoactive intestinal peptide in irritable bowel syndrome
with diarrhea. Scand J Gastroenterol 49(1):43–51
Sprouse-Blum AS, Smith G, Sugai D, Parsa FD (2010) Understanding endorphins and their
importance in pain management. Hawaii Med J 69(3):70–71
Stanczyk MA, Livingston KE, Chang L, Weinberg ZY, Puthenveedu MA, Traynor JR (2019) The
δ-opioid receptor positive allosteric modulator BMS 986187 is a G-protein-biased allosteric
agonist. Br J Pharmacol 176(11):1649–1663
Steinhoff MS, von Mentzer B, Geppetti P, Pothoulakis C, Bunnett NW (2014) Tachykinins and
their receptors: contributions to physiological control and the mechanisms of disease. Physiol
Rev 94(1):265–301
Strand MC, Vindenes V, Gjerde H, Mørland JG, Ramaekers JG (2019) A clinical trial on the acute
effects of methadone and buprenorphine on actual driving and cognitive function of healthy
volunteers. Br J Clin Pharmacol 85(2):442–453
576 A. M. Gantait et al.
Abstract
In the last decades, gasotransmitters have gained attention for their crucial role
in pathophysiological and cellular functions. Gasotransmitters are gaseous
mediators of the cellular signaling and biological responses from one end to the
other. The isoform of these gaseous signaling molecules includes NO (nitric
oxide), CO (carbon monoxide), and H2S (hydrogen sulphide), collectively called
gasotransmitters. The diverse role of these gasotransmitters in cell and molecular
biology as well as biochemical processing has been well validated through
several scientific and clinical studies. The biosynthesis, interaction, and move-
ment of gasotransmitters inside the cellular systems are critical especially in terms
of their pharmacological response. These gaseous molecules are very toxic and
hazardous to human health at higher concentrations but at lower levels they may
be considered as therapeutic agents. They can easily diffuse through all the cell
membrane and act on their targets for generating pharmacological responses. Due
to its gaseous nature, the cellular interactions at the target sites are complex and
make it a critical task for researchers to understand. The mode of action and
molecular pathways are still under the exploratory phase. Apart from their toxic
nature, there are several pharmacological activities such as cardioprotective,
R. Deshmukh · R. K. Harwansh
Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
N. Bandyopadhyay
Molecular Biology Division, National Institute of Malarial Research (NIMR), Dwarka, Delhi, India
S. Bandopadhyay (*)
Faculty of Pharmacy, Naraina Vidya Peeth Group of Institutions, Kanpur, Uttar Pradesh, India
P. Kumar
Department of Pharmacology, Central University of Punjab, Bathinda, Punjab, India
Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical
University, Bathinda, Punjab, India
Keywords
Natural gases · Novel bioactive molecules · ENOG · Signal transduction · Heme
oxygenase · Biomarker
Abbreviations
AD Alzheimer’s Disease
ARDS Acute respiratory distress syndrome
BP Blood pressure
C/EBP CCAATT-enhancer-binding protein
CaM Calcium-binding messenger protein calmodulin
cGMP 3,5-Cyclic guanosine monophosphate
CO Carbon monoxide
CO2 Carbon dioxide
CoHb Carboxyhemoglobin
COPD Chronic obstructive pulmonary disease
CORMs CO releasing molecules
CP Cisplatin
CVD Cardiovascular disease
CYT2E1 Cytochrome P450 2E
DCM Dichloromethane
ED Erectile dysfunction
EDHF Endothelium-derived hyper polarizing factor
EDRF Endothelium-derived relaxing factor
ENOG European Network on Gasotransmitters
eNOS Endothelial NOS
ER Endoplasmic reticulum
GTN Glyceryltrinitrate
GTP Guanosine5-triphosphate
GTT Glucose tolerance test
H 2S Hydrogen sulfide
Hb Hemoglobin
HIF Hypoxia inducible factor
HO Heme oxygenase
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 581
17.1 Introduction
Environmental gases like hydrogen sulfide (H2S), carbon monoxide (CO), carbon
dioxide (CO2), nitric oxide (NO), etc. are treated as toxicants and health hazards.
Apart from being hazardous to health, studies have shown these natural gases have
some health benefits (through cellular and molecular mechanisms), following which
they are considered gasotransmitters. The term gasotransmitter was postulated in the
early twenty-first century after their biological importance in our body as gaseous
signaling molecules was recognized. These signaling molecules became the thrust
area of research to establish their molecular pathways at the cellular level during the
last decade. The human health benefits of gasotransmitters have been explored
through scientific investigation worldwide (Wang 2014; Sukmansky and Reutov
2016; Kolluru et al. 2017).
The fact about these gasotransmitters is that they are highly toxic at higher
concentration but at low concentration they may work as a gaseous signaling
molecule and produce specific biological activities (Andreadou et al. 2015). The
biological role of NO was first discovered in the year 1987, after the identification of
582 R. Deshmukh et al.
These concepts definitely help understand the relevance of NO, CO, H2S, and
others in signal transduction in the human body. They could be novel therapeutic
lead molecules for treatment of several human health problems through scientific
validation. Reportedly, gasotransmitters can effortlessly move across biological
membranes by interacting with a hydrophobic layer of the membrane. For example,
in an animal study it has been observed that normally gaseous molecules move
through biological membranes by using transporter proteins like aquaporin-1. It can
help low molecular weight gases such as NO, CO2 and NH3 to cross cell membranes
easily (Garcia-Mata and Lamattina 2013). The overview of biosynthesis of
gasotransmitters along with their role on vascular system is schematically
represented in Fig. 17.1.
This chapter discusses the role, pathophysiological consequences, biochemistry,
and clinical application of gasotransmitters, particularly that of NO and CO. It will
help us understand the importance of these gasotransmitters as therapeutic bioactive
molecules or drug candidates in association with already established drugs for
enhancing their efficacy against various diseases.
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 583
ENZYME SUBSTRATE
L-arginine Heme L-cysteine
GASOTRANSMITTERS
NO CO H2 S
In the recent past, the importance of different gaseous signaling molecules to their
target sites and related biological responses have been clearly defined and validated
through integrated/scientific approaches. There are various types of gaseous
molecules like NO, CO, CO2, H2S, and so on, which have been proven as gaseous
signaling molecules by transduction the information from one cell to other cells as
biological responses which are endogenously produced (Szabo 2010; Ryter and
Choi 2013; Paul and Snyder 2018a, b). The basic features of gasotransmitters and
neurotransmitters have been explained in Table 17.1.
NO was recognized as the first endogenously produced gaseous molecule. In
various mammal cells, NO was synthesized from L-arginine as substrate by different
analogs of nitric oxide synthase (NOS). Discovery of NO as a novel biological
The ENOG has two major goals for research and regulation of gasotransmitters in
European countries. The first goal is to enhance research on different gaseous
molecules such as NO, CO, H2S, etc. in order to improve their competitiveness,
quality, and efficacy in the development of therapeutic agent against various
diseases. The dissemination of information on gasotransmitters among the
European groups has been through various modes like research problems, skills,
knowledge sharing, expertise, and gasotransmitter biosynthesis. The interactions of
gasotransmitters with their targets for producing cellular effects are also described by
the ENOG among their team. ENOG also help to share the information in multidis-
ciplinary research group through updating and providing the idea, knowledge and
training about gasotransmitters through synthetic chemistry, computational drug
design and pharmacology approaches for establishing gasotransmitters as drug
candidates (Papapetropoulos et al. 2015).
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 585
Table 17.2 Function of ENOG working groups for promotion and development of
gasotransmitters
ENOG groups Functions
Group 1 Molecular control of gasotransmitter production and signaling
Group 2 Gasotransmitters in disease
Group 3 Chemistry and in vitro pharmacology of gasotransmitter-modifying molecules
Group 4 Evaluation of gasotransmitter-modifying agents in animal models of disease
The different ENOG working groups for different purposes are mentioned in
Table 17.2.
17.4.1 Biosynthesis of NO
H
H 2N N OH
NH O2 1M NADPH O
N G-Hydroxyl- L-Arginine
NH2
H
H2N N OH
N O
O2 0.5M NADPH
HO
Citrulline NH2
H
H2 N N OH
O O
NO.
Nitric Oxide
eNOS forms NO in the blood vessel lining that results in vasodilation of smooth
muscle through activation of sGC and cGMP (Wen et al. 2018). It leads to the
constant flow of blood and regulation of blood pressure. Moreover, NO has a
significant role as an anticoagulant by inhibiting platelet and clot formation. But
nNOS-mediated NO has a crucial role in neuronal cell signaling process. In case of
iNOS, it is broadly distributed in the body and contributes in an intrinsic immune
system of the body. During microbial infection/invasion, NOS enzymes are highly
expressed in smooth muscle, macrophages and hepatocyets, resulting in the release
of NO at the microgram level. NO is also found in infected cells along with ROS
(reactive oxygen species) like peroxide (O2), superoxide (S•), and hydroxyl (OH•)
588 R. Deshmukh et al.
radical. It has a certain role in reducing the microbial loads through natural immune
response systems (Doroszko et al. 2018).
Biological activities mainly depend on different NOS enzymes and their location.
There are three subtypes of NOS in mammals: (1) eNOS is responsible for CVS,
(2) nNOS is accountable for the brain or nervous system, and (3) iNOS is liable for
innate immune response (Lee et al. 2017). The NO has a specific role in CVS
through Ca2+-dependent NOS. There are several chemical substances like acetyl-
choline, ATP, bradykinin, and other agents that can stimulate the flux of calcium
known as NOS agonists and can modulate the NO synthesis in vascular endothelial
cells. Likewise, physical factor/agonists, viz., electrical current, light, electromag-
netic fields, acupuncture, flow, and shear stress can stimulate the NO synthesis in
vascular endothelial tissues (Arzumanian et al. 2003; Piazza et al. 2018).
About 90% consumption of NO is taken up by the blood, resulting in inhibition of
platelet and thrombus formation. The rest of the NO diffuses to the vein and arteries
(smooth muscle), and activates a cascade of effects like smooth muscle relaxation.
These effects are directly proportional to the vasodilation that results in reduced
blood pressure. The CVS sustains the NO level along with blood flow. The level of
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 589
NO is dependent upon the rate of blood flow. As the blood flow increases, endothe-
lial tissues increasingly secrete NO to attain a constant level in the blood stream
(Riddell and Owen 1999; Vanhoutte and Gao 2013).
In case of pathological events like atherosclerosis, the NO level is reduced due to
blockade of cholesterol, resulting in vasoconstriction (Li et al. 2014). This abnor-
mality of CVS leads to decreased blood flow and causes increased blood pressure
(hypertension) (Forstermann et al. 2017).
In this context, nitroglycerin is generally used for cardiac disease like hyperten-
sion, which undergoes metabolic transformation and forms NO in CVS endothelial
tissues where it produces vasorelaxation and increases the blood flow in smooth
muscle including atherosclerotic arteries, thereby improving cardiovascular
functions. The NO has a certain role as an anticoagulant by preventing platelet and
blood clot formation in cardiac arteries. If absent there may be chances of coronary
thrombosis, which is a main cause of heart stroke. Thus, NO has a potent gaseous
signaling molecule with an important role in human life (Rochette et al. 2013; Zang
et al. 2018).
The irregular NO generation via different NOS enzymes or signaling cells are related
to several pathological conditions in the organs. In many disease states, NO
metabolites like peroxyl nitrite are raised. The role of NO in various diseases is
discussed in the following.
Adela et al. (2015) reported that the high level of NO was noted with increased
concentration of glucose in serum, which may be due to the activation of endothelial
cell and thereby enhanced NO production. Thus, hyperglycemia enhances NO
generation in Type-2 diabetes (Adela et al. 2015). Moreover, NO exhibits an
important role in the progression and regulation of vascular disease (diabetes and
its complication), which is generally affected by the ROS and NOS (van den Born
et al. 2016).
17.4.4.7 Inflammation
There is a inherent relation between NO mediated apoptosis and inflammation which
in turns depend upon the concentration level at the target site. iNOS produced higher
concentrations of NO cause apoptosis (a programmed cell death) while eNOS and
nNOS mediated lower level of NO can produce cytoprotective activity. For example
ONOO species which may be considered for apoptotic determining process,
although the specific function of ONOO in inflammatory cell apoptosis is still
un-cleared. Hence the concept of NO-apoptosis is applicable in inflammatory
conditions (Lee et al. 2017).
It has been studied that activated macrophages triggers the sensitive tumors
(murine) and thus initiating NO based apoptosis in both activated anti-tumor
T cells and normal tumor cells. Therefore, NO mediated macrophage induces
apoptosis (death of nearby cells) that raises the elimination of apoptotic cells results
in facilitating the resolution phase of inflammatory environment. These inflamed
cells lead to cancerous due to oxidant enriched surroundings, after sometimes these
phenomena cause relentless and self-responsive oxidative stress due to generation of
reactive oxygen species (ROS) and reactive nitrogen species (RNS) (MacMicking
et al. 1997a, b; Baran et al. 2004).
17.4.4.8 Cancer
In negative perspective, some of the NO related activities is related to oxidative
stress in various cancers including melanoma. At low level of NO (<100 nM), it can
produce the metastasis, proliferation and angiogenesis while at higher levels of
400–500 nM it may cause cytotoxicty and cell death (apoptosis). Therefore, NO
donors have been considered as a promising treatment approach of different cancers.
NO-NSAIDs are good example of this approach that NO-NSAIDs have been
reported to be effective in arresting the metastasis and tumor proliferation as
compared to pure drug itself (Abdelall et al. 2017).
NO shows anticancer property against colon cancer (HCT116 cancer cell) by
inhibiting cell proliferation and production (Olah et al. 2018). In case of melanoma,
NO mediates its effect via enzymatic process and free radical production. Thereby,
NO can help in formation and proliferation of melanoma by a bunch of mechanisms
involving inhibition of apoptosis and immune cells, alteration of angiogenic
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 593
17.4.4.9 Tuberculosis
iNOS-based NO has a crucial role in host defense systems against Mycobacterium
tuberculosis in tuberculosis (TB) infection. TB has an internal complex
environments to combat the therapeutic agent, resist and counter the effects of
NO, RNS and ROS in intracellular surrounding of bacteria. The bacterial
proteosome in TB is responsible for resistance against host-mediated nitrosative
and oxidative stress by deprivation of stress-induced protein damage. It has been
reported that TB genes (noxR1 and noxR3) have protection ability against oxidative
molecules (ROS, RNS, and NO) and their harmful events, i.e., apoptosis and
necrosis (Bhat et al. 2017; Braverman and Stanley 2017a, b).
The TB bacteria can change nitrates to nitrites in case of hypoxia, which favors
anaerobic environment and dormancy stage for TB. Recent reports suggest that
NO is an important gas transporter for host defense mechanism and diversified
pathophysological consequences of Mycobacterium tuberculosis infections. NO
could be explored along with anti-tubercular drugs for enhancement of their efficacy.
The quest for developing highly effective anti-tubercular agents and vaccines are
utmost required for prevention of TB (Chinta et al. 2016a, b).
mice. Evidence suggests for establishing new concept for understanding of the
molecular mechanism of NO with endothelial impairment in the context of cognitive
decline and AD progression. Moreover, endothelial NO is recognized as a protector
of healthy mind and key agent establishing activity between CNS and cerebrovas-
cular system (Katusic and Austin 2014; Austin and Katusic 2016).
17.5.1 Biosynthesis of CO
Fig. 17.3 Biosynthesis of CO by hemoglobin degradation mechanism of red blood cells catalyzed
by heme oxygenase (HO) (Adapted from Fig. 2 of Wu and Wang 2005a, b)
which is excreted from the body via urinary pathway (Fig. 17.3). During injury, lysis
of RBC takes place and deoxygenated Hb is formed, which is observed as a dark
red/purple color formation. Later on a green tinge appears at injury site due to
oxidation of heme to form biliverdin. This pigment is gradually converted to
bilirubin causing a yellow coloration (Johnson et al. 2003).
HO is reported to have three isoforms, i.e., inducible HO-1 and two constitutive
forms, namely, HO-2 and HO-3. The HO-1 has low levels of expression in most
tissues under physiologic conditions except spleen. HO-1, known as the stress
protein (i.e., hsp32) is a redox-sensitive response protein. HO-I activity is
up-regulated in oxidative stress like inflammatory reaction or disease-like
conditions. Furthermore, HO-1 is up-regulated in case of cellular stress and therefore
found to be a vital antioxidant enzyme. Depending on the nature of the stimuli and
cell types, the induction of HO-1 can be moderated by various signaling pathways
like protein kinase C (PKC), cAMP-dependent mechanisms, Ca2+-calmodulin-
dependent protein kinase and the phosphoinositol pathways (Durante et al. 1997;
Immenschuh et al. 1998; Terry et al. 1999; Wu and Wang 2006). In addition, it has
been reported that mitogen-activated protein kinases (ERK and P38) and tyrosine
phosphorylation induce HO-1 in some tissues (Alam et al. 2000; Chen and Maines
2000).
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 597
The catabolism of the CO takes place by three mechanisms, i.e., expiration, scav-
enging, and oxidation, which are discussed below.
17.5.2.1 Expiration
CO is majorly expelled from the systemic production via lungs. CO diffuses through
the alveolar-capillary membrane, which is mainly dependent on the amount of
hemoglobin in the pulmonary capillaries and alveolar gas volume (Untereiner
et al. 2012). The concentration of CO in the alveoli is determined by the partial
598 R. Deshmukh et al.
pressure of oxygen and CO as these two gases compete for binding of same iron site.
Nowadays, CO is used to measure the heme metabolism and level of bilirubin.
Moreover, the level of CO is also used clinically for the determination of jaundice in
infants (Stevenson et al. 1994; Okuyama et al. 2001).
17.5.2.2 Scavenging
In normal physiological conditions, HO catalyzed heme degradation mechanism is
the major source of endogenously generated CO in our body. The body store of CO
is a combination of CO inhaled from the environment and xenobiotics metabolized
by hepatocytes (Kubic and Anders 1978). About 80% of CO binds with Hb present
in RBC and forms as COHb and other heme proteins which are accountable for the
remaining CO load (Coburn 1970). The CO body stores are transferable. During
hypoxia, it is reported that CO moves from blood to tissue where CO binds to heme
proteins (Coburn and Mayers 1971).
17.5.2.3 Oxidation
It has been believed that oxidation of CO results in the formation of CO2 involving
cytochrome c oxidase of mitochondria. However, no studies have been reported in
mammalian tissues related to its oxidation and that too under physiological
conditions. The microbes that live in CO oxidize it to form CO2 in the presence of
CO dehydrogenase. Moreover, in chemistry the oxidation or of CO to CO2 is also
well established. Also the atmospheric CO reacts with hydroxyl radicals to yield
HO2 (hydroperoxyl radical) and CO2 (Allen and Root 1957).
Anti-Apoptotic Effects
CO exhibits apoptosis in various cells and tissues of the body in a very selective and
specific manner. For instance, it shows an anti-apoptotic to hepatocytes,
cardiomyocytes, and endothelial cells, thus protecting the cells and tissues from
injury. The anti-apoptotic activity of CO is dependent on p38 MAPK signaling,
phosphorylation of protein kinase R (e.g., endoplasmic reticulum kinase), and/or
through Akt activation (Choi et al. 2003; Clark et al. 2003; Kim et al. 2008). Besides,
CO also inhibits tumor necrosis factor-(TNF) -α- and endoplasmic reticulum
(ER) stress-induced cellular apoptosis. Interestingly, mitochondrial membrane
permeabilization can be prevented by low CO concentrations (10–100 μM) in
isolated mouse liver cells, thus stopping the formation of pro-apoptotic molecules.
However, high level of CO (250–500 μM) can activate the swelling in mitochondria
(Queiroga et al. 2011).
Pro-Apoptotic Effects
CO exhibits pro-apoptotic effect on fibroblasts and hyper-proliferative smooth
muscle cells (Zheng et al. 2009). It also blocks the smooth muscle cells of the
trachea in humans and propagation of vascular smooth muscle cell in rats both under
hypoxic and normoxic states. Zhou et al. showed an inhibition of fibroblast prolifer-
ation with the decrease degree of fibrosis in mice when exposed to CO. They
discovered that CO blocked the cell cycle in G0/G1 phase via a cGMP-dependent
mechanism. CO at a concentration of 100–200 ppm inhibited the proliferation of
VSMC at G(1)/S transition phase and obstructed the activation and production of
cyclin A. Thus, the HO/CO signaling system offers an important function in the
regulation of cell survival (Zhou et al. 2005).
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 601
17.5.4.1 Diabetes
Diabetes is a metabolic disease distinguished by high of glucose in the systemic
circulation. It can be classified as insulin-dependent (Type 1) or non-insulin-depen-
dent (Type 2) diabetes mellitus. It has been observed that in diabetic patients there is
an increase exhaled CO levels. The CO concentration in normal healthy persons is
2.9 ppm whereas it is observed to be at 4.0 and 5.0 ppm for Type 1 and Type
2, respectively. Thus, there exists a relationship between glycemia and exhaled CO
levels. This observation was further validated through oral GTT (glucose tolerance
test). In case of healthy humans, there is a connection between blood glucose
concentrations (i.e., 3.9–5.5 mM) and amount of CO expired (i.e., 3.0–6.3 ppm).
This abnormal level is restored to normal within 40 min of glucose intake (Paredi
et al. 1999). This relationship was explained by Lundquist and associate in intact
mouse islets wherein HO activity was initiated by glucose. In an experiment with
Goto-Kakizaki rats model (i.e., defective pancreatic β-cell with HO-2 expression),
low CO was observed with insulin deficiency. This study signified that HO-2 has an
important function in insulin secretion and glucose metabolism (Henningsson et al.
1999).
Hypertension
Hypertension is termed as consistent increased blood pressure distinguished by
increased systolic pressure (i.e., above 150 mm Hg) and high diastolic pressure
(i.e., above 90 mm Hg). It is characterized by vascular structural changes like
atherosclerosis (hardening of blood vessels due to fat deposition) and co-arctation
of aorta (narrowing of the aorta) which results in high resistance and elevated blood
pressure. This hemodynamic stress causes initiation of HO-1 activity, which in turn
stimulates the activity of sGC, and cGMP in VSMCs (Wegiel et al. 2010).
CO at a concentration of 60 ppm can decrease the hypertrophy of left ventricles
and aorta in mouse model which results in the inhibition of angiotensin II-dependent
hypertension. CO can reduce the phosphorylation NOX and Akt resulting in reduc-
ing ROS production, thereby protecting the cells (Ndisang et al. 2004).
Pulmonary arterial hypertension (PAH) is typified as high blood pressure of
lungs. The obstruction in small arteries in the lung causes increased pressure in the
vessels leading to right ventricle failure. CO has a capacity to reduce the PAH.
602 R. Deshmukh et al.
Kobayashi et al. demonstrated that in mice the PAH and right ventricular hypertro-
phy are reversed by inhalation of CO (250 ppm, 1 h/day for 2 or 3 weeks). Moreover,
it also restores the pressure in the pulmonary artery and right ventricule along with
pulmonary vascular structural design (Kobayashi et al. 2007).
The underlying principle in the inhibition of hypertension is the involvement of
the NOS pathway. The HO-1/CO system can be up-regulated by hemin supplemen-
tation, which can be used in the treatment of hypertension. In a study in young
hypertensive rats, hemin therapy was reported to decrease the blood pressure. A 21-
days-based implanted hemin osmotic minipumps releasing 15 mg/kg/day provide
prolonged safety against hypertension in 12-week-old SHRs. It is the accumulation
of residual hemin within VSMCs that results in normalization of blood pressure of
SHR. Thus, this new approach of hemin application at physiologically or therapeutic
concentrations can be a novel alternative approach for the management of
hypertension.
Atherosclerosis
Atherosclerosis is a disease characterized by the hardening of the arteries due to the
deposition of plaque. It slowly and silently blocks arteries, obstructing the blood
flow in the heart. It is one of the primary causes of strokes, heart attacks, and
peripheral vascular disease. It involves inflammation, endothelial dysfunction, and
vascular proliferation. It is evident that during atherosclerosis, HO-1 is up-regulated,
which is important in the management of disease (Johnson et al. 2006).
CO acts as strong anti-atherosclerotic agent owing to VSMCs apoptotic
properties, initiation of endothelial cell proliferation, and anti-inflammatory activity.
Wang et al. reported that in balloon angioplasty-induced vessel injury in rats, CO at a
concentration of 250 ppm blocked the development of atherosclerotic lesions. The
CO blocks leukocyte infiltration and VSMC proliferation thereby resulting in pro-
tection of arteries from hyperplasia (Wang et al. 2001).
Ion channels are the vital constituent of a cell, which is responsible for the activity of
any pharmaceuticals. The activity of CO also depends on its interaction with various
ion channels, such as K+ channels. The K+ channels superfamily is composed of
calcium-activated KCa channels, voltage-dependent Kv, and ATP-sensitive KATP.
In the late 1990s, the concept of delivering low concentrations of CO gas had
begun being investigated for alleviating various pathophysiological conditions
characterized by oxidative stress and inflammatory states. This was a new and
quite provocative proposal concept for management of pathology of disease. How-
ever, it is also evident that CO interferes in oxygen transport and its delivery.
Therefore, its sustained inhalation can cause problems which are related to systemic
effects and thus limiting the use of CO in therapeutics. Later on it was found that this
drawback could be overcome by packing CO in a more stable chemical form called
CORMs. The different types of CO and their properties are given in Table 17.3.
Initially, CO was considered a toxic gas and a “by-product” of heme metabolism
in human physiology. Only recently, the pharmacology of CO has been explored in
human physiology and recognized as a gasotransmitter. CO and CORMs are now
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 605
O O
17.5.6.1 Inflammation
CO and CORMs can be used in modulating immune-suppression and inflammation.
They act as therapeutic agents in numerous models of inflammatory disease such
as COPD, asthma, and airway hyper-responsiveness, and carrageenan-induced
mesenteric inflammation. In addition, many phase II clinical trials have also
validated the anti-inflammatory activity of these gasotransmitters. CORM-2 can be
employed in the management of allergy as it significantly lowers the release of
histamine and expression of CD203c in the mast cells of guinea pigs and human
basophils (Vannacci et al. 2004).
The lipopolysaccharide (LPS)-activated murine J774 macrophagecells,
stimulates the inflammatory response by the production of NO and TNF- α involving
NF-κB. CORM-2 inhibits the translocation of NF-κB into the nucleus and
completely eliminates the inflammatory response. Also, CORM-2 reduces the
expression of inflammatory molecules like nitrite and iNOS in LPS stimulated
macrophages (Lee et al. 2003). In a Plasmodium infected mouse experimental
cerebral malaria model where the infection stimulated an inflammation, the inhala-
tion of CO for 3 days after the infection considerably reduced the inflammation in
606
R. Deshmukh et al.
Fig. 17.4 Summary of Therapeutic Applications of CO and CORMs (Adapted from Fig. 2 of Ling et al. 2017)
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 607
mouse brain and improved the survival rate; in contrast there were heavy casualties
in the control group (Pamplona et al. 2007).
17.5.6.5 Cancer
Intriguingly, in cancer therapy HO-1/CO has dichotomous function. At low
concentrations, HO-1/CO, in cancer cells exerts cytoprotective, pro-proliferative
and pro-angiogenic property. However, at high concentration, CO exhibits antitumor
activity. HO-1 overexpression has been established in several tumor cells such as
pancreatic cancer, prostate cancer, melanoma, and Kaposi sarcoma. Generally, CO at
the physiological dose can be formed via tumor cells or tumor-infiltrating
macrophages to produce a pro-tumor effect. Therefore, inhibition of HO-1 can be
utilized for anticancer therapy. For example, in a mouse cancer model small
interfering RNA (siRNA) mediated HO-1 silencing results in lower tumor growth
rates and angiogenesis. Inhaled CO or CORMs at a higher concentration could be
lethal to tumor cells (Simon et al. 2011).
In a CD1 athymic mouse model inhalation of 500 ppm CO for 1 h a day
considerably lowers the cancer cell growth and the resultant angiogenesis. The
anti-tumor activity might be due to mitochondrial ROS generation, acceleration of
oxygen consumption, inhibition of cellular protein synthesis, and reduction of
cellular antioxidants. The high level of CO is toxic to the normal tissue; therefore,
these preclinical data may not be clinically valid (Wegiel et al. 2013).
17.6 Conclusions
References
Abdelall EKA, Abdelhamid AO, Azouz AA (2017) Synthesis and biological evaluations of new
nitric oxide-anti-inflammatory drug hybrids. Bioorg Med Chem Lett 27(18):4358–4369
Abraham NG, Kappas A (2008) Pharmacological and clinical aspects of heme oxygenase.
Pharmacol Rev 60:79–127
Abraham NG, Jiang H, Balazy M, Goodman AI (2003) Methods for measurements of heme
oxygenase (HO) isoforms-mediated synthesis of carbon monoxide and HO-1 and HO-2
proteins. Methods Mol Med 86:399–411
Adela R, Nethi SK, Bagul PK, Barui AK, Mattapally S, Kuncha M, Patra CR, Reddy PN, Banerjee
SK (2015) Hyperglycaemia enhances nitric oxide production in diabetes: a study from south
Indian patients. PLoS One 10(4):e0125270
Akter F, Coghlan G, de Mel A (2016) Nitric oxide in paediatric respiratory disorders: novel
interventions to address associated vascular phenomena? Ther Adv Cardiovasc Dis 10
(4):256–270
Al Omar S, Salama H, Al HM, Al RH, Bunahia M, El Kasem W, Siddiqui FJ, Dilawar M, Yassin H,
Masud F, Mohamed A, Mansour A (2016) Effect of early adjunctive use of oral sildenafil and
inhaled nitric oxide on the outcome of pulmonary hypertension in newborn infants. A feasibility
study. J Neonatal Perinatal Med 9(3):251–259
Alam J, Wicks C, Stewart D, Gong P, Touchard C, Otterbein L, Choi A, Burow M, Tou J.-s. (2000)
Mechanism of Heme Oxygenase-1 gene activation by cadmium in MCF-7 mammary epithelial
cells. Role of OF p38 kinase and Nrf2 transcription factor. J Biol Chem 275(36):27694–27702
Allen TA, Root WS (1957) Partition of carbon monoxide and oxygen between air and whole blood
of rats, dogs and men as affected by plasma pH. J Appl Physiol 10(2):186–190
610 R. Deshmukh et al.
Amsel SDJ, Kevin J, Soden KJ, Sielken RL Jr, Valdez-Flora C (2001) Observed versus predicted
carboxyhemoglobin levels in cellulose triacetate workers exposed to methylene chloride. Am J
Ind Med 40(2):180–191
Andreadou I, Iliodromitis EK, Rassaf T, Schulz R, Papapetropoulos A, Ferdinandy P (2015) The
role of gasotransmitters NO, H2S and CO in myocardial ischaemia/reperfusion injury and
cardioprotection by preconditioning, postconditioning and remote conditioning. Br J Pharmacol
172(6):1587–1606
Antosova M, Mokra D, Pepucha L, Plevkova J, Buday T, Sterusky M, Bencova A (2017)
Physiology of nitric oxide in the respiratory system. Physiol Res 66(Supplementum 2):S159–
S172
Archakov AI, Karuzina II, Petushkova N, Lisitsa AV, Zgoda V (2002) Production of carbon
monoxide by cytochrome P450 during iron-dependent lipid peroxidation. Toxicol In Vitro 16
(1):1–10
Arzumanian V, Stankevicius E, Laukeviciene A, Kevelaitis E (2003) Mechanisms of nitric oxide
synthesis and action in cells. Medicina (Kaunas) 39(6):535–541
Austin SA, Katusic ZS (2016) Loss of endothelial nitric oxide synthase promotes p25 generation
and tau phosphorylation in a murine model of Alzheimer’s disease. Circ Res 119
(10):1128–1134
Bagul A, Hosgood S, Kaushik M, Nicholson ML (2008) Carbon monoxide protects against
ischemia-reperfusion injury in an experimental model of controlled nonheartbeating donor
kidney. Transplantation 85(4):576–581
Baran CP, Zeigler MM, Tridandapani S, Marsh CB (2004) The role of ROS and RNS in regulating
life and death of blood monocytes. Curr Pharm Des 10(8):855–866
Basudhar D, Somasundaram V, de Oliveira GA, Kesarwala A, Heinecke JL, Cheng RY, Glynn SA,
Ambs S, Wink DA, Ridnour LA (2017) Nitric oxide Synthase-2-derived nitric oxide drives
multiple pathways of breast Cancer progression. Antioxid Redox Signal 26(18):1044–1058
Bhat KH, Srivastava S, Kotturu SK, Ghosh S, Mukhopadhyay S (2017) The PPE2 protein of
Mycobacterium tuberculosis translocates to host nucleus and inhibits nitric oxide production.
Sci Rep 7:39706
Boehning D, Moon C, Sharma S, Hurt KJ, Hester LD, Ronnett GV, Shugar D, Snyder SH (2003)
Carbon monoxide neurotransmission activated by CK2 phosphorylation of heme oxygenase-2.
Neuron 40(1):129–137
Bohlen HG (2015) Nitric oxide and the cardiovascular system. Compr Physiol 5(2):808–823
Botros FT, Navar LG (2006) Interaction between endogenously produced carbon monoxide and
nitric oxide in regulation of renal afferent arterioles. Am J Phys Heart Circ Phys 291(6):H2772–
H2778
Braverman J, Stanley SA (2017a) Nitric oxide modulates macrophage responses to Mycobacterium
tuberculosis infection through activation of HIF-1alpha and repression of NF-kappaB. J
Immunol 199(5):1805–1816
Braverman J, Stanley SA (2017b) Nitric oxide modulates macrophage responses to Mycobacterium
tuberculosis infection through activation of HIF-1alpha and repression of NF-kappaB. J
Immunol 199(5):1805–1816
Burnett AL (2006) The role of nitric oxide in erectile dysfunction: implications for medical therapy.
J Clin Hypertens (Greenwich) 8(12 Suppl 4):53–62
Cartledge J, Minhas S, Eardley I (2001) The role of nitric oxide in penile erection. Expert Opin
Pharmacother 2(1):95–107
Cepinskas G, Katada K, Bihari A, Potter RF (2008) Carbon monoxide liberated from carbon
monoxide-releasing molecule CORM-2 attenuates inflammation in the liver of septic mice.
Am J Physiol Gastrointest Liver Physiol 294(1):G184–G191
Chen K, Maines MD (2000) Nitric oxide induces heme oxygenase-1 via mitogen-activated protein
kinases ERK and p38. Cell Mol Biol (Noisy-le-Grand) 46(3):609–617
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 611
Chen B, Guo L, Fan C, Bolisetty S, Joseph R, Wright MM, Agarwal A, George JF (2009) Carbon
monoxide rescues Heme Oxygenase-1-deficient mice from arterial thrombosis in allogeneic
aortic transplantation. Am J Pathol 175(1):422–429
Cheng Y, Rong J (2017) Therapeutic potential of Heme Oxygenase-1/carbon monoxide system
against ischemia-reperfusion injury. Curr Pharm Des 23(26):3884–3898
Chin BY, Jiang G, Wegiel B, Wang HJ, MacDonald T, Zhang XC, Gallo D, Cszimadia E, Bach FH,
Lee PJ, Otterbein LE (2007) Hypoxia-inducible factor 1α stabilization by carbon monoxide
results in cytoprotective preconditioning. Proc Natl Acad Sci 104(12):5109–5114
Chinta KC, Saini V, Glasgow JN, Mazorodze JH, Rahman MA, Reddy D, Lancaster JR Jr, Steyn AJ
(2016a) The emerging role of gasotransmitters in the pathogenesis of tuberculosis. Nitric Oxide
59:28–41
Chinta KC, Saini V, Glasgow JN, Mazorodze JH, Rahman MA, Reddy D, Lancaster JR Jr, Steyn AJ
(2016b) The emerging role of gasotransmitters in the pathogenesis of tuberculosis. Nitric Oxide
59:28–41
Choi YK (2018) Role of carbon monoxide in neurovascular repair processing. Biomol Ther (Seoul)
26(2):93–100
Choi BM, Pae HO, Kim YM, Chung HT (2003) Nitric oxide-mediated cytoprotection of
hepatocytes from glucose deprivation-induced cytotoxicity: involvement of heme oxygenase-
1. Hepatology 37(4):810–823
Cifuentes D, Poittevin M, Bonnin P, Ngkelo A, Kubis N, Merkulova-Rainon T, Levy BI (2017)
Inactivation of nitric oxide synthesis exacerbates the development of Alzheimer disease pathol-
ogy in APPPS1 mice (amyloid precursor protein/Presenilin-1). Hypertension 70(3):613–623
Clark JE, Naughton P, Shurey S, Green CJ, Johnson TR, Mann BE, Foresti R, Motterlini R (2003)
Cardioprotective actions by a water-soluble carbon monoxide-releasing molecule. Circ Res 93
(2):e2–e8
Coburn RF (1970) The carbon monoxide body stores. Ann N Y Acad Sci U S A 174(1):11–22
Coburn RF, Mayers LB (1971) Myoglobin O2 tension determined from measurement of
carboxymyoglobin in skeletal muscle. Am J Phys 220(1):66–74
Coburn RF, Blakemore WS, Forster RE (1963a) Endogenous carbon monoxide production in man.
J Clin Invest 42:1172–1178
Coburn RF, Blakemore WS, Forster RE (1963b) Endogenous carbon monoxide production in man.
J Clin Invest 42:1172–1178
Csongradi E, Juncos LA, Drummond HA, Vera T, Stec DE (2012) Role of carbon monoxide in
kidney function: is a little carbon monoxide good for the kidney? Curr Pharm Biotechnol 13
(6):819–826
Dallas ML, Scragg JL, Peers C (2009) Inhibition of L-type Ca(2+) channels by carbon monoxide.
Adv Exp Med Biol 648:89–95
Dashwood MR, Crump A, Shi-Wen X, Loesch A (2011) Identification of neuronal nitric oxide
synthase (nNOS) in human penis: a potential role of reduced neuronally-derived nitric oxide in
erectile dysfunction. Curr Pharm Biotechnol 12(9):1316–1321
Divakaran S, Loscalzo J (2017) The role of nitroglycerin and other nitrogen oxides in cardiovascu-
lar therapeutics. J Am Coll Cardiol 70(19):2393–2410
Doroszko A, Dobrowolski P, Radziwon-Balicka A, Skomro R (2018) New insights into the role of
oxidative stress in onset of cardiovascular disease. Oxidative Med Cell Longev 2018:9563831
Durante W, Christodoulides N, Cheng K, Peyton KJ, Sunahara RK, Schafer AI (1997) CAMP
induces heme oxygenase-1 gene expression and carbon monoxide production in vascular
smooth muscle. Am J Physiol 273(1 Pt 2):H317–H323
Fabiani J-N, Achouh PE, Simonet S, Verbeuren TJ (2008) Carbon monoxide induces relaxation of
human internal thoracic and radial arterial grafts. Interact Cardiovasc Thorac Surg 7(6):959–962
Fadel PJ (2017) Nitric oxide and cardiovascular regulation: beyond the endothelium. Hypertension
69(5):778–779
Farrugia G, Szurszewski JH (2014) Carbon monoxide, hydrogen sulfide, and nitric oxide as
signaling molecules in the gastrointestinal tract. Gastroenterology 147(2):303–313
612 R. Deshmukh et al.
Fleissner F, Thum T (2011) Critical role of the nitric oxide/reactive oxygen species balance in
endothelial progenitor dysfunction. Antioxid Redox Signal 15(4):933–948
Fordjour PA, Wang Y, Shi Y, Agyemang K, Akinyi M, Zhang Q, Fan G (2015) Possible
mechanisms of C-reactive protein mediated acute myocardial infarction. Eur J Pharmacol
760:72–80
Foresti R, Hammad J, Clark J, Johnson TR, Mann BE, Friebe A, Green C, Motterlini R (2004)
Vasoactive properties of CORM-3, a novel water-soluble carbon monoxide-releasing molecule.
Br J Pharmacol 142(3):453–460
Forstermann U (2010) Nitric oxide and oxidative stress in vascular disease. Pflugers Arch 459
(6):923–939
Forstermann U, Sessa WC (2012) (2012). Nitric oxide synthases: regulation and function. Eur Heart
J 33(7):829–837
Forstermann U, Xia N, Li H (2017) Roles of vascular oxidative stress and nitric oxide in the
pathogenesis of atherosclerosis. Circ Res 120(4):713–735
Freitas A, Alves-Filho JC, Secco DD, Neto AF, Ferreira SH, Barja-Fidalgo C, Cunha FQ (2006)
Heme oxygenase/carbon monoxide-biliverdin pathway down regulates neutrophil rolling, adhe-
sion and migration in acute inflammation. Br J Pharmacol 149(4):345–354
Fujimoto H, Ohno M, Ayabe S, Kobayashi H, Ishizaka N, Kimura H, Yoshida K-I, Nagai R (2004)
Carbon monoxide protects against cardiac ischemia – reperfusion injury in vivo via MAPK and
Akt – eNOS pathways. Arterioscler Thromb Vasc Biol 24(10):1848–1853
Fujisaki N, Kohama K, Nishimura T, Yamashita H, Ishikawa M, Kanematsu A, Yamada T, Lee S,
Yumoto T, Tsukahara K, Kotani J, Nakao A (2016) Donor pretreatment with carbon monoxide
prevents ischemia/reperfusion injury following heart transplantation in rats. Med Gas Res 6
(3):122–129
Garcia-Mata C, Lamattina L (2013) Gasotransmitters are emerging as new guard cell signaling
molecules and regulators of leaf gas exchange. Plant Sci 201-202:66–73
Hayashi S, Omata Y, Sakamoto H, Higashimoto Y, Hara T, Sagara Y, Noguchi M (2004)
Characterization of rat heme oxygenase-3 gene. Implication of processed pseudogenes derived
from heme oxygenase-2 gene. Gene. https://doi.org/10.1016/j.gene.2004.04.002
Heinemann SH, Hoshi T, Westerhausen M, Schiller A (2014) Carbon monoxide--physiology,
detection and controlled release. Chem Commun (Camb) 50(28):3644–3660
Heneka MT, Kummer MP, Stutz A, Delekate A, Schwartz S, Vieira-Saecker A, Griep A, Axt D,
Remus A, Tzeng TC, Gelpi E, Halle A, Korte M, Latz E, Golenbock DT (2013) NLRP3 is
activated in Alzheimer’s disease and contributes to pathology in APP/PS1 mice. Nature 493
(7434):674–678
Henningsson R, Alm P, Ekström P, Lundquist I (1999) Heme oxygenase and carbon monoxide:
regulatory roles in islet hormone release - A biochemical, immunohistochemical, and confocal
microscopic study. Diabetes 48(1):66–76
Hettiarachchi N, Dallas M, Al-Owais M, Griffiths H, Hooper N, Scragg J, Boyle J, Peers C (2014)
Heme oxygenase-1 protects against Alzheimer’s amyloid-beta(1-42)-induced toxicity via car-
bon monoxide production. Cell Death Dis 5:e1569
Hou S, Xu R, Heinemann SH, Hoshi T (2008) The RCK1 high-affinity Ca2+ sensor confers carbon
monoxide sensitivity to Slo1 BK channels. Proc Natl Acad Sci U S A. https://doi.org/10.1073/
pnas.0800304105
Immenschuh S, Kietzmann T, Hinke V, Wiederhold M, Katz N, Muller-Eberhard U (1998) The rat
heme oxygenase-1 gene is transcriptionally induced via the protein kinase A signaling pathway
in rat hepatocyte cultures. Mol Pharmacol 53:483–491
Johnson TR, Mann BE, Clark J, Foresti R, Green C, Motterlini R (2003) Metal carbonyls: a new
class of pharmaceuticals? Angew Chem Int Ed Engl 42(32):3722–3729
Johnson F, Johnson RA, Durante W, Jackson K, Stevenson BK, Peyton KJ (2006) Metabolic
syndrome increases endogenous carbon monoxide production to promote endothelial dysfunc-
tion in obese Zucker rats. Am J Physiol Regul Integr Comp Physiol 290(3):R601–R608
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 613
Katusic ZS, Austin SA (2014) Endothelial nitric oxide: protector of a healthy mind. Eur Heart J 35
(14):888–894
Keshet R, Erez A (2018) Arginine and the metabolic regulation of nitric oxide synthesis in cancer.
Dis Model Mech 11(8):dmm033332
Kim HS, Loughran PA, Rao J, Billiar TR, Zuckerbraun BS (2008) Carbon monoxide activates
NF-kappaB via ROS generation and Akt pathways to protect against cell death of hepatocytes.
Am J Physiol Gastrointest Liver Physiol 295(1):G146–G152
Kobayashi A, Ishikawa K, Matsumoto H, Kimura S, Kamiyama Y, Maruyama Y (2007) Synergetic
antioxidant and vasodilatory action of carbon monoxide in angiotensin II - induced cardiac
hypertrophy. Hypertension 50(6):1040–1048
Kolluru GK, Shen X, Yuan S, Kevil CG (2017) Gasotransmitter heterocellular signaling. Antioxid
Redox Signal 26(16):936–960
Kovacevic Z, Sahni S, Lok H, Davies MJ, Wink DA, Richardson DR (2017) Regulation and control
of nitric oxide (NO) in macrophages: protecting the “professional killer cell” from its own
cytotoxic arsenal via MRP1 and GSTP1. Biochim Biophys Acta Gen Subj. 1861(5 Pt
A):995–999
Kubic VL, Anders MW (1978) Metabolism of dihalomethanes to carbon monoxide—III: studies on
the mechanism of the reaction. Biochem Pharmacol 27(19):2349–2355
Lakkisto P, Kytö V, Forsten H, Siren J-M, Segersvärd H, Voipio-Pulkki L-M, Laine M, Pulkki K,
Tikkanen I (2010) Heme oxygenase-1 and carbon monoxide promote neovascularization after
myocardial infarction by modulating the expression of HIF-1α, SDF-1α and VEGF-B, vol
635, pp 156–164
Lamon BD, Zhang FF, Puri N, Brodsky SV, Goligorsky MS, Nasjletti A (2009) Dual pathways of
carbon monoxide–mediated vasoregulation. Circ Res 105(8):775–783
Lee T-S, Tsai H-L, Chau L-Y (2003) Induction of heme oxygenase-1 expression in murine
macrophages is essential for the anti-inflammatory effect of low dose 15-deoxy-Δ12,14-prosta-
glandin J2. J Biol Chem 278:19325–19330
Lee M, Rey K, Besler K, Wang C, Choy J (2017) Immunobiology of nitric oxide and regulation of
inducible nitric oxide synthase. Results Probl Cell Differ 62:181–207
Leffler CW, Nasjletti A, Yu C, Johnson RA, Fedinec AL, Walker N (1999) Carbon monoxide and
cerebral microvascular tone in newborn pigs. Am J Phys Heart Circ Phys 276(5):H1641–H1646
Li VG, Sacerdoti D, Sangras B, Vanella A, Mezentsev A, Scapagnini G, Falck J, Abraham NG
(2005) Carbon monoxide signaling in promoting angiogenesis in human microvessel endothe-
lial cells. Antioxid Redox Signal. https://doi.org/10.1089/ars.2005.7.704
Li A, Xi Q, Umstot ES, Bellner L, Schwartzman ML, Jaggar JH, Leffler CW (2008) Astrocyte-
derived CO is a diffusible messenger that mediates glutamate-induced cerebral arteriolar dilation
by activating smooth muscle cell KCa channels. Circ Res 102(2):234–241
Li L, Hsu A, Moore PK (2009) Actions and interactions of nitric oxide, carbon monoxide and
hydrogen sulphide in the cardiovascular system and in inflammation—a tale of three gases!
Pharmacol Ther. https://doi.org/10.1016/j.pharmthera.2009.05.005
Li H, Horke S, Forstermann U (2014) Vascular oxidative stress, nitric oxide and atherosclerosis.
Atherosclerosis 237(1):208–219
Lin HH, Lai SC, Chau LY (2011) Heme oxygenase-1/carbon monoxide induces vascular endothe-
lial growth factor expression via p38 kinase-dependent activation of Sp1. J Biol Chem 286
(5):3829–3838
Ling K, Men F, Wang W-C, Zhou Y-Q, Zhang H-W, Ye D-W (2017) Carbon monoxide and its
controlled release: therapeutic application, detection, and development of carbon monoxide
releasing molecules (CORMs). J Med Chem. https://doi.org/10.1021/acs.jmedchem.6b01153
Lowenstein CJ, Dinerman JL, Snyder SH (1994) Nitric oxide: a physiologic messenger. Ann Intern
Med 120(3):227–237
Luo S, Lei H, Qin H, Xia Y (2014) Molecular mechanisms of endothelial NO synthase uncoupling.
Curr Pharm Des 20(22):3548–3553
614 R. Deshmukh et al.
MacMicking J, Xie QW, Nathan C (1997a) Nitric oxide and macrophage function. Annu Rev
Immunol 15:323–350
MacMicking J, Xie QW, Nathan C (1997b) Nitric oxide and macrophage function. Annu Rev
Immunol 15:323–350
McCoubrey WK Jr, Huang TJ, Maines MD (1997) Isolation and characterization of a cDNA from
the rat brain that encodes hemoprotein heme oxygenase-3. Eur J Biochem 247(2):725–732
Miller MR, Megson IL (2007) Recent developments in nitric oxide donor drugs. Br J Pharmacol
151(3):305–321
Mir JM, Maurya R (2018) A gentle introduction to gasotransmitters with special reference to nitric
oxide: biological and chemical implications. Rev Inorg Chem 38(4):193–220
Moncada S, Higgs A (1993) The L-arginine-nitric oxide pathway. N Engl J Med 329
(27):2002–2012
Motterlini R, Foresti R (2017) Biological signaling by carbon monoxide and carbon monoxide-
releasing molecules. Am J Physiol Cell Physiol 312(3):C302–C313
Motterlini R, Otterbein LE (2010) The therapeutic potential of carbon monoxide. Nat Rev Drug
Discov 9(9):728–743
Munzel T, Daiber A (2018) Inorganic nitrite and nitrate in cardiovascular therapy: A better
alternative to organic nitrates as nitric oxide donors? Vasc Pharmacol 102:1–10
Nagpure BV, Bian JS (2016) Interaction of hydrogen sulfide with nitric oxide in the cardiovascular
system. Oxidative Med Cell Longev 2016:6904327
Nakao A, Huang C-S, Stolz DB, Wang Y, Franks J, Tochigi N, R Billiar T, Toyoda Y, Tzeng E, R
McCurry K (2011) Ex vivo carbon monoxide delivery inhibits intimal hyperplasia in arterialized
vein grafts. Cardiovasc Res. https://doi.org/10.1093/cvr/cvq298
Naseem KM (2005) The role of nitric oxide in cardiovascular diseases. Mol Asp Med 26
(1–2):33–65
Ndisang JF, Tabien HEN, Wang R (2004) Carbon monoxide and hypertension. J Hypertens 22
(6):1057–1074
Neilly PJ, Kirk SJ, Gardiner KR, Rowlands BJ (1994) The L-arginine/nitric oxide pathway--
biological properties and therapeutic applications. Ulster Med J 63(2):193–200
Nizamutdinova IT, Kim YM, Kim HJ, Seo HG, Lee JH, Chang KC (2009) Carbon monoxide (from
CORM-2) inhibits high glucose-induced ICAM-1 expression via AMP-activated protein kinase
and PPAR-γ activations in endothelial cells. Atherosclerosis 207(2):405–411
Nobre LS, Seixas JD, Romão CC, Saraiva LM (2007) Antimicrobial action of carbon monoxide-
releasing compounds. Antimicrob Agents Chemother 51(12):4303–4307
Okuyama H, Yonetani M, Uetani Y, Nakamura H (2001) End-tidal carbon monoxide is predictive
for neonatal non-hemolytic hyperbilirubinemia. Pediatr Int 43(4):329–333
Olah G, Modis K, Toro G, Hellmich MR, Szczesny B, Szabo C (2018) Role of endogenous and
exogenous nitric oxide, carbon monoxide and hydrogen sulfide in HCT116 colon cancer cell
proliferation. Biochem Pharmacol 149:186–204
Omar SA, Webb AJ, Lundberg JO, Weitzberg E (2016) Therapeutic effects of inorganic nitrate and
nitrite in cardiovascular and metabolic diseases. J Intern Med 279(4):315–336
Owens EO (2010) Endogenous carbon monoxide production in disease. Clin Biochem 43
(15):1183–1188
Pamplona A, Ferreira A, Balla J, Jeney V, Balla G, Epiphanio S, Chora Â, Rodrigues CD,
Grégoire I, Cunha-Rodrigues M, Portugal S, P Soares M, Mota M (2007) Heme oxygenase-1
and carbon monoxide suppress the pathogenesis of experimental cerebral malaria. Nat Med. 13
(6):703–710
Papapetropoulos A, Foresti R, Ferdinandy P (2015) Pharmacology of the ‘gasotransmitters’ NO,
CO and H2S: translational opportunities. Br J Pharmacol 172(6):1395–1396
Paredi P, Biernacki W, Invernizzi G, Kharitonov SA, Barnes PJ (1999) Exhaled carbon monoxide
levels elevated in diabetes and correlated with glucose concentration in blood: A new test for
monitoring the disease? Chest 116(4):1007–1011
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 615
Paul BD, Snyder SH (2018a) Gasotransmitter hydrogen sulfide signaling in neuronal health and
disease. Biochem Pharmacol 149:101–109
Paul BD, Snyder SH (2018b) Gasotransmitter hydrogen sulfide signaling in neuronal health and
disease. Biochem Pharmacol 149:101–109
Piazza M, Dieckmann T, Guillemette JG (2018) Investigation of the structure and dynamic of
calmodulin-nitric oxide synthase complexes using NMR spectroscopy. Front Biosci (Landmark
Ed) 23:1902–1922
Pitsikas N (2015) The role of nitric oxide in the object recognition memory. Behav Brain Res
285:200–207
Polhemus DJ, Lefer DJ (2014) Emergence of hydrogen sulfide as an endogenous gaseous signaling
molecule in cardiovascular disease. Circ Res 114(4):730–737
Poulos TL, Li H (2017) Nitric oxide synthase and structure-based inhibitor design. Nitric Oxide
63:68–77
Queiroga C, Almeida A, Alves P, Brenner C, Vieira H (2011) Carbon monoxide prevents hepatic
mitochondrial membrane permeabilization. BMC Cell Biol 12:10
Quillon A, Fromy B, Debret R (2015) Endothelium microenvironment sensing leading to nitric
oxide mediated vasodilation: a review of nervous and biomechanical signals. Nitric Oxide
45:20–26
Raman K, Barbato JE, Ifedigbo E, Ozanich BA, Zenati M, Otterbein L, Tzeng E (2006) Inhaled
carbon monoxide inhibits intimal hyperplasia and provides added benefit with nitric oxide. J
Vasc Surg. 44(1):151–158
Ramlawi B, Scott JR, Feng J, Mieno S, Raman KG, Gallo D, Csizmadia E, Yoke CB, Bach FH,
Otterbein LE, Sellke FW (2007) Inhaled carbon monoxide prevents graft-induced intimal
hyperplasia in swine. J Surg Res 138(1):121–127
Riddell DR, Owen JS (1999) Nitric oxide and platelet aggregation. Vitam Horm 57:25–48
Rochette L, Lorin J, Zeller M, Guilland JC, Lorgis L, Cottin Y, Vergely C (2013) Nitric oxide
synthase inhibition and oxidative stress in cardiovascular diseases: possible therapeutic targets?
Pharmacol Ther 140(3):239–257
Ryter SW, Choi AM (2013) Carbon monoxide: present and future indications for a medical gas.
Korean J Intern Med 28(2):123–140
Shahrbaf MA, Mahjoob MP, Khaheshi I, Akbarzadeh MA, Barkhordari E, Naderian M, Tajrishi FZ
(2018) The role of air pollution on ST-elevation myocardial infarction: A narrative mini review.
Futur Cardiol 14(4):301–306
Shefa U, Yeo SG, Kim MS, Song IO, Jung J, Jeong NY, Huh Y (2017) Role of gasotransmitters in
oxidative stresses, neuroinflammation, and neuronal repair. Biomed Res Int. https://doi.org/10.
1155/2017/1689341. Epub 2017 Mar 12
Simon T, Anegon I, Blancou P (2011) Heme oxygenase and carbon monoxide as an immunothera-
peutic approach in transplantation and cancer. Immunotherapy 3(4 Suppl):15–18
Sjostrand T (1952) The formation of carbon monoxide by the decomposition of haemoglobin
in vivo. Acta Physiol Scand 26(4):338–344
Socco S, Bovee RC, Palczewski MB, Hickok JR, Thomas DD (2017) Epigenetics: the third pillar of
nitric oxide signaling. Pharmacol Res 121:52–58
Stein A, Guo Y, Tan W, Wu W-J, Zhu X, Li Q, Luo C, Dawn B, Johnson TR, Motterlini R, Bolli R
(2005) Administration of a CO-releasing molecule induces late preconditioning against
myocardial infarction. J Mol Cell Cardiol 38(1):127–134
Stevenson DK, Vreman HJ, Oh W, Fanaroff AA, Wright LL, Lemons JA, Verter J, Shankaran S,
Tyson JE, Korones SB et al (1994) Bilirubin production in healthy term infants as measured by
carbon monoxide in breath. Clin Chem 40(10):1934–1939
Stewart RD, Fisher TN, Hosko MJ, Peterson JE, Baretta ED, Dodd HC (1972) Carboxyhemoglobin
elevation after exposure to dichloromethane. Science 176(4032):295–296
Sukmansky OI, Reutov VP (2016) Gasotransmitters: physiological role and involvement in the
pathogenesis of the diseases. Usp Fiziol Nauk 47(3):30–58
616 R. Deshmukh et al.
Szabo C (2010) Gaseotransmitters: new frontiers for translational science. Sci Transl Med 2
(59):59ps54. https://doi.org/10.1126/scitranslmed.3000721
Takehito T, Yoshifumi M (1988) Metabolism of dichloromethane and the subsequent binding of its
product, carbon monoxide, to cytochrome P-450 in perfused rat liver. Toxicol Lett 40(1):93–96
Tayem Y, Johnson TR, Mann BE, Green CJ, Motterlini R (2006) Protection against cisplatin-
induced nephrotoxicity by a carbon monoxide-releasing molecule. Am J Physiol Renal Physiol
290(4):F789–F794
Tenhunen R, Marver HS, Schmid R (1968) The enzymatic conversion of heme to bilirubin by
microsomal heme oxygenase. Proc Natl Acad Sci U S A 61(2):748–755
Terry C, Clikeman JA, Hoidal JR, Callahan KS (1999) TNF-alpha and IL-1alpha induce heme
oxygenase-1 via protein kinase C, Ca2+, and phospholipase A2 in endothelial cells. Am J
Physiol 276(5):H1493–H1501
Thom SR (1990) Carbon monoxide-mediated brain lipid peroxidation in the rat. J Appl Physiol 68
(3):997–1003
Traub O, Van Bibber R (1995) Role of nitric oxide in insulin-dependent diabetes mellitus-related
vascular complications. West J Med 162(5):439–445
Truss NJ, Warner T (2011a) Gasotransmitters and platelets. Pharmacol Ther 132(2):196–203
Truss NJ, Warner TD (2011b) Gasotransmitters and platelets. Pharmacol Ther 132(2):196–203
Untereiner A, Wu L, Wang R (2012) The role of carbon monoxide as a gasotransmitter. In:
Hermann A, Sitdikova G, Weiger T (eds) Gasotransmitters: physiology and pathophysiology,
pp 37–70
Urquhart P, Rosignoli G, Cooper D, Motterlini R, Perretti M (2007) Carbon monoxide-releasing
molecules modulate leukocyte-endothelial interactions under flow. J Pharmacol Exp Ther 321
(2):656–662
van den Born JC, Hammes HP, Greffrath W, van Goor H, Hillebrands JL, DFG GRK International
Research Training Group 1874 Diabetic Microvascular Complications (DIAMICOM) (2016)
Gasotransmitters in vascular complications of diabetes. Diabetes 65(2):331–345
Van Landeghem L, Laleman W, Van der Elst I, Zeegers M, Van Pelt J, Cassiman D, Nevens F
(2009) Carbon monoxide produced by intrasinusoidally located haem-oxygenase-1 regulates
the vascular tone in cirrhotic rat liver. Liver Int 29(5):650–660
Vanhoutte PM, Gao Y (2013) Beta blockers, nitric oxide, and cardiovascular disease. Curr Opin
Pharmacol 13(2):265–273
Vannacci A, Di Felice A, Giannini L, Marzocca C, Pierpaolo S, Zagli G, Masini E, Mannaioni PF
(2004) The effect of carbon monoxide releasing molocule on the immunological activation of
guinea-pig mast cells and human basophils. Inflamm Res; 53 Suppl 1:S9–S10
Varadi J, Lekli I, Juhasz B, Bácskay I, Szabo G, Gesztelyi R, Szendrei L, Varga E, Bak I, Foresti R,
Motterlini R, Tosaki A (2007) Beneficial effects of carbon monoxide-releasing molecules on
post-ischemic myocardial recovery, vol 80, pp 1619–1626
Venketaraman V, Talaue MT, Dayaram YK, Peteroy-Kelly MA, Bu W, Connell ND (2003) Nitric
oxide regulation of L-arginine uptake in murine and human macrophages. Tuberculosis (Edinb)
83(5):311–318
Verma A, Hirsch DJ, Glatt CE, Ronnett GV, Snyder SH (1993) Carbon monoxide: a putative neural
messenger. Science 259(5093):381–384
Von Burg R (1999) Carbon monoxide. J Appl Toxicol 19(5):379–386
Wagner CT, Durante W, Christodoulides N, Hellums JD, Schafer AI (1997) Hemodynamic forces
induce the expression of heme oxygenase in cultured vascular smooth muscle cells. J Clin Invest
100(3):589–596
Wallace JL, Ianaro A, Flannigan KL, Cirino G (2015) Gaseous mediators in resolution of inflam-
mation. Semin Immunol 27(3):227–233
Wang R (1998) Resurgence of carbon monoxide: an endogenous gaseous vasorelaxing factor. Can J
Physiol Pharmacol 76(1):1–15
Wang R (2014) Gasotransmitters: growing pains and joys. Trends Biochem Sci 39(5):227–232
17 Pharmacology of Gasotransmitters (Nitric Oxide and Carbon Monoxide). . . 617
Wang R, Wu L, Wang Z (1997) The direct effect of carbon monoxide on KCa channels in vascular
smooth muscle cells. Pflugers Arch 434(3):285–291
Wang R, Wang Z, Wu L, Hanna ST, Peterson-Wakeman R (2001) Reduced vasorelaxant effect of
carbon monoxide in diabetes and the underlying mechanisms. Diabetes 50(1):166–174
Wang Z, Wu Y, Zhang S, Zhao Y, Yin X, Wang W, Ma X, Liu H (2019) The role of NO-cGMP
pathway inhibition in vascular endothelial-dependent smooth muscle relaxation disorder of
AT1-AA positive rats: protective effects of adiponectin. Nitric Oxide 87:10–22
Wegiel B, Gallo DJ, Raman KG, Karlsson JM, Ozanich B, Chin BY, Tzeng E, Ahmad S, Ahmed A,
Baty CJ, Otterbein LE (2010) Nitric oxide–dependent bone marrow progenitor mobilization by
carbon monoxide enhances endothelial repair after vascular injury. Circulation 121(4):537–548
Wegiel B, Gallo D, Csizmadia E, Harris C, Belcher J, Vercellotti GM, Penacho N, Seth P,
Sukhatme V, Ahmed A, Pandolfi PP, Helczynski L, Bjartell A, Persson JL, Otterbein LE
(2013) Carbon monoxide expedites metabolic exhaustion to inhibit tumor growth. Cancer Res
73(23):7009–7021
Wen L, Feil S, Wolters M, Thunemann M, Regler F, Schmidt K, Friebe A, Olbrich M, Langer H,
Gawaz M, de Wit C, Feil R (2018) A shear-dependent NO-cGMP-cGKI cascade in platelets acts
as an auto-regulatory brake of thrombosis. Nat Commun 9(1):4301
Wobst J, Kessler T, Dang TA, Erdmann J, Schunkert H (2015) Role of sGC-dependent NO
signalling and myocardial infarction risk. J Mol Med (Berl) 93(4):383–394
Wu L, Wang R (2005a) Carbon monoxide: endogenous production, physiological functions, and
pharmacological applications. Pharmacol Rev 57(4):585–630
Wu L, Wang R (2005b) Carbon monoxide: endogenous production, physiological functions, and
pharmacological applications. Pharmacol Rev 57(4):585–630
Wu L, Wang R (2006) Carbon monoxide: endogenous production, physiological functions, and
pharmacological applications. Pharmacol Rev 57(4):585–630
Xi Q, Tcheranova D, Parfenova H, Horowitz B, Leffler CW, Jaggar JH (2004) Carbon monoxide
activates KCa channels in newborn arteriole smooth muscle cells by increasing apparent Ca2+
sensitivity of alpha-subunits. Am J Physiol Heart Circ Physiol 286(2):H610–H618
Yarlagadda K, Hassani J, Foote IP, Markowitz J (2017) The role of nitric oxide in melanoma.
Biochim Biophys Acta Rev Cancer 1868(2):500–509
Zang Y, Popat KC, Reynolds MM (2018) Nitric oxide-mediated fibrinogen deposition prevents
platelet adhesion and activation. Biointerphases 13(6):06E403
Zhao Y, Vanhoutte PM, Leung SW (2015) Vascular nitric oxide: beyond eNOS. J Pharmacol Sci
129(2):83–94
Zheng L, Zhou Z, Lin L, Alber S, Watkins S, Kaminski N, Choi AMK, Morse D (2009) Carbon
monoxide modulates α–smooth muscle actin and small proline rich-1a expression in fibrosis.
Am J Respir Cell Mol Biol 41(1):85–92
Zhou Z, Song R, Fattman C, Greenhill S, Alber S, Oury T, Choi A, Morse D (2005) Carbon
monoxide suppresses bleomycin-induced lung fibrosis, vol 166, pp 27–37
Sodium Channels: As an Eye of the Storm
in Various Clinical Pathologies 18
Vinod Tiwari, Ankit Uniyal, Akhilesh, Anagha Gadepalli,
Vineeta Tiwari, and Somesh Agrawal
Abstract
Voltage-gated sodium ion channels are essential to maintain the excitability and
activity of neurons and neuronal network. Several studies have been done to
explore the basic properties of ion channels, their existence and physiological
characteristics. Till date we know that 11 genes are responsible for encoding of
9 families of sodium channels (Nav 1.1 to Nav 1.9) and are classified according to
varying degrees of sensitivity to Tetrodotoxin (TTX). These are localized in
various sites such as skeletal muscles, central nervous system (CNS), cardiac
muscles, and peripheral sensory neurons. Any aberration in its structure and
function leads to various clinical pathologies such as channelopathies (where
dysregulation in receptors are directly responsible for initiation and progression)
and diseases contributing to dysregulation of expression of sodium channels
cause various neurological disorders. In this chapter, we emphasize the composi-
tion, function, and regulation of sodium channels at the molecular level and the
crucial role of sodium channels in the development and progression of various
disease pathologies such as epilepsy, schizophrenia, familiar hemiplegic migraine
and neuropathic pain.
Keywords
Abbreviations
18.1 Introduction
Voltage-gated sodium ion channels are required for excitation of cells. Their history
can be traced back to 1941, when Kenneth S. Cole and Richard F. Baker confirmed
the existence of voltage-gated membrane pores in giant squid axon (Cole and Curtis
1939). Alan Hodgkin and Andrew Huxley won the Nobel prize for their work on
basic properties of ion channels and their mechanisms of excitation and inhibition in
1952. Later in 1970 and 1981, the existence and physiological characteristics of ion
channels were explored by Bernard Katz, Ricardo, and Miledi et al. and Erwin Neher
and Bert Sakmann et al., using noise analysis and patch clamp techniques, respec-
tively (Cole and Baker 1941; Roger et al. 2015).
So far, 11 genes are responsible for the encoding of 9 families of sodium channels
(Nav 1.1 to Nav 1.9). These genes are classified according to varying degrees of
sensitivity to Tetrodotoxin (TTX) and can be traced to four paralogous locations in
the chromosome segment (Ambrose et al. 1992; Wang et al. 1992; Burgess et al.
1995; Beckers et al. 1996; Kozak and Sangameswaran 1996; Plummer et al. 1998;
Catterall et al. 2005; Bagal et al. 2015) (Table 18.1). More than 20 exons encode for
9 sodium channel α-subunits. Nav channels (1.1, 1.2, 1.3, and 1.7) are situated on the
second chromosome in both humans as well as in the mouse whereas Nav channels
1.5 are situated at 2q24 and in case of mouse, present on chromosome 2. And the
Nav 1.8 and 1.9 channels are present on 3p21–24 in case of humans and chromo-
some 9 in the mouse. Skeletal muscle contains the Nav 1.4 and the CNS contains the
18 Sodium Channels: As an Eye of the Storm in Various Clinical Pathologies 621
Table 18.1 Classification of voltage-gated sodium channels based on location and tetrodotoxin
sensitivity (in human and mouse)
Nav Location
channel Tetrodotoxin
subtype Human Mouse sensitivity References
Nav 1.1 Chromosome Chromosome Sensitive Catterall et al. (2005)
2 2
Nav 1.2 Chromosome Chromosome Sensitive Catterall et al. (2005)
2 2
Nav 1.3 Chromosome Chromosome Sensitive Catterall et al. (2005)
2 2
Nav 1.4 Chromosome Chromosome Sensitive Ambrose et al. (1992); Catterall
17 11 et al. (2005); Wang et al. (1992)
Nav 1.5 2q 24 Chromosome Resistant Catterall et al. (2005)
2
Nav 1.6 Chromosome Chromosome Sensitive Burgess et al. (1995); Catterall
12 15 et al. (2005); Plummer et al.
(1998)
Nav 1.7 Chromosome Chromosome Sensitive Beckers et al. (1996); Catterall
2 2 et al. (2005); Kozak and
Sangameswaran (1996)
Nav 1.8 3p 21–24 Chromosome Resistant Catterall et al. (2005)
9
Nav 1.9 3p 21–24 Chromosome Resistant Catterall et al. (2005)
9
Nav 1.6. The genes of Nav 1.4 are situated on the 17th chromosome (humans) and
11th chromosome (mouse) whereas genes of Nav 1.6 can be traced to the 12th
chromosome (humans) and the 15th chromosome (mouse) (Catterall et al. 2005;
Mantegazza and Catterall 2012). All the sodium channels structures are analogous to
each other, but few amino acid replacements confer this resistance, for example, in
Nav 1.5 which is predominantly localized in cardiac muscles, substitution of phe-
nylalanine to cystine in pore area provides 200 times reduction in sensitivity to TTX
(Yamagishi et al. 2001). A similar replacement is also observed in Nav 1.8 and Nav
1.9, which are localized in peripheral sensory neurons where phenylalanine is
replaced by serine, which provides even greater resistance (Mantegazza and Catterall
2012). Channelopathies are the group of diseases characterized by any aberration in
the structure or function arising from a mutation in the genes encoding for sodium
channels. In this chapter, we will study the composition, function, and regulation at
the molecular level of sodium channels and the crucial role of sodium channels in the
development and progression of various disease pathologies (Roger et al. 2015).
622 V. Tiwari et al.
α subunit
β subunit
DI D II D III D IV
S S S S S S S S S S S S S S S S S S S S S S S S
12345 6 12345 6 12345 6 12345 6
Alanine Asparagine
IFMT Segment
Fig. 18.1 Composition of sodium channel: α subunit (Domains- I, II, III & IV); β subunit and
IFMT segment (Isoleucine, Phenylalanine, Methionine, and Tryptophan)
Voltage-gated sodium channels (VGSCs) occur in three forms: (1) active (2) refrac-
tory, (3) closed. From closed state to go on to open state requires depolarization,
changes in membrane polarity to positive, which usually happens during an action
potential, and within a few milli-seconds VGSCs progress to refractory state, also
known as the inactive state, is a mechanism that protects cells from excessive
stimulation. This is followed by the closed state when the cell membrane attains
the normal repolarized potential. During closed state, the “voltage sensor” segment
4 is held inside due to the overall negative resting membrane potential of the cell. The
depolarization relieves the electrostatic force on segment 4 pushing it to move out,
thus opening the intracellular ion gate. This allows sodium ion conductions, leading
to increased sodium concentration inside the cell. When the intracellular gate is
opened, the IFMT segment binds to hydrophobic amino acids (like alanine and
asparagine) between segment 4 and segments 5 of DIII and DIV, as there is no steric
hindrance caused by intracellular ion gate due to conformational change. This state is
called a refractory state/inactive state. In the inactive state, sodium ions cannot pass
through causing prevention of the cell from overstimulation. From a refractory state
to go to closed state again requires a change in membrane potential. Whenever cell
resting membrane potential is restored, segment 4 domain is attracted toward the
cell, augmenting its electrostatic influence on segment 4 resulting in a conforma-
tional change to the closed ion channel (Catterall 1992; Marban et al. 1998)
(Fig. 18.2).
Channel Interacting Proteins (ChIPs) are endogenous proteins that interact with
voltage-gated Na+ channels. Some examples of ChIPs are connexion 43, Caveolin
3, Calcium-calmodulin kinase 2, ankyrins, telethonin, plakophilin, neuronal precur-
sor cell-expressed developmentally downregulated 4 (nedd4), fibroblast growth
factor (FGF) and its homologous factors (FHFs). These endogenous proteins regu-
late the sodium channel expression and its functioning (Savio-Galimberti et al.
2012).
624 V. Tiwari et al.
Na+
1.Closed Na+ 2.Open
+
+
+
+
S SSSSS Depolarization S
S S SSS
6 54321 4
6 5 321
+
+
+
+
IFMT IFMT Intracellular ion gate
Intracellular ion gate
3.Inactivated
+
Repolarisation +
S
S S SSS
4
6 5 321
+
+
Na+
+ve RMP ++++++++++++
Fig. 18.2 Molecular functioning of sodium channels: Closed, opened and inactivated state; IFMT
segment (Isoleucine, Phenylalanine, Methionine, and Tryptophan), RMP; Resting Membrane
Potential
neuropathic conditions like Charcot and Marie tooth disorder and neuropathic pain.
Another study proved that β cell stimulation leads to increased surface expression of
VGSCs. This is due to intracellular VGSCs pool mobilization by caveolin 3 and
caveolae membrane proteins, VGSCs are surface expressed (Savio-Galimberti et al.
2012; Roger et al. 2015).
Sodium channel as discussed above is crucial for the functioning of any excitable
cells, and any aberration in its structure or functioning leads to various clinical
pathologies. There are two types of disorders that are classified based on the role
played by the sodium channels on pathogenesis of diseases: (1) channelopathies
(where dysfunctional receptors are directly responsible for initiation and progres-
sion) and (2) disease contributing to dysregulated expression of sodium channels
(like cancer, neurological disorders) (Kaplan et al. 2016).
Sodium channels are important to maintain the excitable character as well as the
activity of neurons and the neuronal network. They are ubiquitously expressed
throughout the neurocyton and axons in neurons. Denseness of sodium channel is
more at Nodes of Ranvier and axon initial segment (AIS), which is responsible for
speeding nerve impulse conduction. This phenomenon is often referred to as salta-
tory conduction (Kaplan et al. 2016). The action potential is initiated from AIS and
this characteristic is attributed to relatively high sodium channel receptor density at
this site. Various IHC studies have validated this claim of determining distribution of
sodium channel expression along AIS. In such studies Nav1.6 was densely
expressed at distal AIS of retinal ganglion cell of rat, whereas Nav1.1 was
aggregated at proximal AIS. Similar trend in expression was observed in cortical
pyramidal neurons of rat showing increased Nav1.2 and Nav 1.6 at AIS. Hyperpo-
larization of NaV1.6 that depend on NaV 1.2 activity results in differential expres-
sion of sodium channel. When depolarlizing signals enter AIS, activation threshold
of Nav 1.6 crosses more readily as compared to Nav1.2, which consequently
generates action potential. This causes two types of currents: the first one propagates
down the axon and the other results in backpropagating currents, which leads to
activation of Nav1.2. This action potential is propagated from soma to axons and
dendrites. The backpropagating currents are crucial for relating the neuronal output
and regulating neuronal platicity. Dendritic VGSCs also contributes to action poten-
tial by amplifying the depolarlization response. Collectively, this data suggests the
complex relationship of sodium channels distribution and neuronal activity. The data
also hints at the susceptibility of brain networks to any aberrations in sodium
channels function and its number by mutations, disorders, or action of drug (Kaplan
et al. 2016).
626 V. Tiwari et al.
Analysis of mutation in VGSCs in epilepsy has shown a complex data that results in
a specific pattern showing connection between channel mutation and epilepsy. In
VGSCs of interneuron, there are alterations in channel biological organization that
are cell dependent in nature; for example, there is a loss of function in Nav1.1 or
Nav1.6 due to depolarization in shifts in the voltage dependence of activation,
hyperpolarization in shifts in the voltage dependence of inactivation, and
haploinsufficiency (Hargus et al. 2013). Contrastingly, alteration in pyramidal cell
channels in NaV1.2 or NaV1.6 leads to depolarization of shifts in the voltage
dependence of inactivation and augmentation in persistent current (Ye et al. 2018).
Haploinsufficiency in SCNIA triggers Dravet Syndrome (Bechi et al. 2012) and
functional dropping in SCN1B (Patino et al. 2009). The function in SCN2A is
associated with BFNIS (Misra et al. 2008). The functional gaining in SCN8A is
related to epileptic encephalopathy and functional loss is related with its nonoccur-
rence (Martin et al. 2007). In patients of epilepsy, additional potential pathological
mechanisms have been suggested, but not yet specifically identified. Mutations in
the voltage sensor in votage-gated sodium channel result in exchange of the amino,
acid, i.e., arginines (charged) with residues (neutral). As a result, voltage sensor
leads to development of a leak current (omega/gating pore current) (Sokolov et al.
2005). In spite of the fact that it is not seen in epilepsy patients till now, in
hypokalemic periodic paralysis, Nav1.4 mutation is associated with an increase in
omega current, proposing involvement in channelopathies (Sokolov et al. 2007).
Resurgence of sodium current is proposed to be involved in epileptical conditions. In
some subtypes of voltage-gated sodium channels, by membrane repolarization
followed by prolonged depolarization pulse, a minor transient ingoing current can
be attained (Raman and Bean 1997). Augmented resurgent current amplitude has
18 Sodium Channels: As an Eye of the Storm in Various Clinical Pathologies 627
attainment of function and was associated with FHM as well as increased occurrence
of generalized epilepsy (Kahlig et al. 2008).
disorder is manifested by extreme pain and burning sensation in rectal, ocular and
submandibular region, which causes change in depolarizing voltage, which in turn
effects steady-state inactivation.
18.4 Conclusion
Normal functional and number of sodium channels are crucial for the functioning of
any excitable cells. This, when mutations or disorder arise, which disturbs this
balance, leads to various clinical pathologies. Channelopathies are disarrays arising
18 Sodium Channels: As an Eye of the Storm in Various Clinical Pathologies 631
that are able to target these markers can be developed or repurposed as an alternative
safe therapy for treatment of channelopathies.
References
Ambrose C et al (1992) The α-subunit of the skeletal muscle sodium channel is encoded proximal to
Tk-1 on mouse chromosome 11. Mamm Genome 3:151–155. https://doi.org/10.1007/
BF00352459
Bagal SK et al (2015) Voltage gated sodium channels as drug discovery targets. Channels
9:360–366. https://doi.org/10.1080/19336950.2015.1079674
Bechi G et al (2012) Pure haploinsufficiency for Dravet syndrome Na V1.1 (SCN1A) sodium
channel truncating mutations. Epilepsia 53:87–100. https://doi.org/10.1111/j.1528-1167.2011.
03346.x
Beckers MC et al (1996) A new sodium channel α-subunit gene (Scn9a) from Schwann cells maps
to the Scn1a, Scn2a, Scn3a cluster of mouse chromosome 2. Genomics 36:202–205. https://doi.
org/10.1006/geno.1996.0447
Black JA et al (2004) Changes in the expression of tetrodotoxin-sensitive sodium channels within
dorsal root ganglia neurons in inflammatory pain. Pain 108:237–247. https://doi.org/10.1016/j.
pain.2003.12.035
Burgess DL et al (1995) Mutation of a new sodium channel gene, Scn8a, in the mouse mutant
“motor endplate disease”. Nat Genet 10:461–465. https://doi.org/10.1038/ng0895-461
Carreño O et al (2013) Screening of CACNA1A and ATP1A2 genes in hemiplegic migraine:
clinical, genetic, and functional studies. Mol Genet Genomic Med 1:206–222. https://doi.org/
10.1002/mgg3.24
Catterall WA (1992) Cellular and molecular biology of voltage-gated sodium channels. Physiol Rev
72:S15–S48. https://doi.org/10.1152/physrev.1992.72.suppl_4.S15
Catterall WA et al (2005) Nomenclature and structure-function relationships of voltage-gated
calcium channels. Pharmacol Rev. https://doi.org/10.1124/pr.57.4.5.units
Cole KS, Baker RF (1941) Longitudinal impedance of the squid giant axon. J Gen Physiol. https://
doi.org/10.1085/jgp.24.6.771
Cole KS, Curtis HJ (1939) Electric impedance of the squid giant axon during activity. J Gen
Physiol. https://doi.org/10.1085/jgp.22.5.649
Cregg R et al (2013) Novel mutations mapping to the fourth sodium channel domain of Nav1.
7 result in variable clinical manifestations of primary erythromelalgia. Neuromol Med 15
(2):265–278. Springer
Dabby R et al (2011) Chronic non-paroxysmal neuropathic pain—novel phenotype of mutation in
the sodium channel SCN9A gene. J Neurol Sci 301(1–2):90–92. Elsevier
Dichgans M et al (2005) Mutation in the neuronal voltage-gated sodium channel SCN1A in familial
hemiplegic migraine. Lancet 366:371–377. https://doi.org/10.1016/S0140-6736(05)66786-4
Frank HY, Catterall WA (2003) Overview of the voltage-gated sodium channel family. Genome
Biol 4(3):207. BioMed Central
Hargus NJ et al (2013) Evidence for a role of Na v 1.6 in facilitating increases in neuronal
hyperexcitability during epileptogenesis. J Neurophysiol 110:1144–1157. https://doi.org/10.
1152/jn.00383.2013
Harty TP, Waxman SG (2007) Inactivation properties of sodium channel Nav1.8 maintain action
potential amplitude in small DRG neurons in the context of depolarization. Mol Pain. https://doi.
org/10.1186/1744-8069-3-12
18 Sodium Channels: As an Eye of the Storm in Various Clinical Pathologies 633
Imbrici P, Camerino DC, Tricarico D (2013) Major channels involved in neuropsychiatric disorders
and therapeutic perspectives. Front Genet 4. https://doi.org/10.3389/fgene.2013.00076
Jarecki BW et al (2010) Human voltage-gated sodium channel mutations that cause inherited
neuronal and muscle channelopathies increase resurgent sodium currents. J Clin Investig
120:369–378. https://doi.org/10.1172/JCI40801
Kahlig KM et al (2008) Divergent sodium channel defects in familial hemiplegic migraine. Proc
Natl Acad Sci 105:9799–9804. https://doi.org/10.1073/pnas.0711717105
Kaplan DI, Isom LL, Petrou S (2016) Role of sodium channels in epilepsy. Cold Spring Harb
Perspect Med 6. https://doi.org/10.1101/cshperspect.a022814
King GF, Vetter I (2014) No gain, no pain: NaV1. 7 as an analgesic target. ACS Chem Neurosci 5
(9):749–751. ACS Publications
Kozak CA, Sangameswaran L (1996) Genetic mapping of the peripheral sodium channel genes,
Scn9a and Scn10a, in the mouse. Mamm Genome 7:787–788. https://doi.org/10.1007/
s003359900235
Lindia JA et al (2005) Relationship between sodium channel NaV1.3 expression and neuropathic
pain behavior in rats. Pain 117:145–153. https://doi.org/10.1016/j.pain.2005.05.027
Lolignier S et al (2011) Nav1.9 channel contributes to mechanical and heat pain hypersensitivity
induced by subacute and chronic inflammation. PLoS One 6:e23083. https://doi.org/10.1371/
journal.pone.0023083
Mantegazza M, Catterall WA (2012) Voltage-gated Na+ channels: structure, function, and patho-
physiology. In: Jasper’s basic mechanisms of the epilepsies. National Center for Biotechnology
Information (US), Bethesda, MD
Marban E, Yamagishi T, Tomaselli GF (1998) Structure and function of voltage-gated sodium
channels. J Physiol 508:647–657. https://doi.org/10.1111/j.1469-7793.1998.647bp.x
Martin MS et al (2007) The voltage-gated sodium channel Scn8a is a genetic modifier of severe
myoclonic epilepsy of infancy. Hum Mol Genet 16:2892–2899. https://doi.org/10.1093/hmg/
ddm248
Misra SN, Kahlig KM, George AL (2008) Impaired NaV1.2 function and reduced cell surface
expression in benign familial neonatal-infantile seizures. Epilepsia 49:1535–1545. https://doi.
org/10.1111/j.1528-1167.2008.01619.x
Nassar MA et al (2006) Nerve injury induces robust allodynia and ectopic discharges in Nav 1.3
null mutant mice. Mol Pain 2:1744-8069-2-33. https://doi.org/10.1186/1744-8069-2-33
Patino GA et al (2009) A functional null mutation of SCN1B in a patient with Dravet syndrome. J
Neurosci 29:10764–10778. https://doi.org/10.1523/JNEUROSCI.2475-09.2009
Plummer NW et al (1998) Exon organization, coding sequence, physical mapping, and polymor-
phic intragenic markers for the human neuronal sodium channel gene SCN8A. Genomics
54:287–296. https://doi.org/10.1006/geno.1998.5550
Raman IM, Bean BP (1997) Resurgent sodium current and action potential formation in dissociated
cerebellar Purkinje neurons. J Neurosci 17(5):4517–4526
Rees E et al (2019) Targeted sequencing of 10,198 samples confirms abnormalities in neuronal
activity and implicates voltage-gated sodium channels in schizophrenia pathogenesis. Biol
Psychiatry 85:554–562. https://doi.org/10.1016/j.biopsych.2018.08.022
Roger S et al (2015) Voltage-gated sodium channels and cancer: is excitability their primary role?
Front Pharmacol 6. https://doi.org/10.3389/fphar.2015.00152
Savio-Galimberti E, Gollob MH, Darbar D (2012) Voltage-gated sodium channels: biophysics,
pharmacology, and related channelopathies. Front Pharmacol 3. https://doi.org/10.3389/fphar.
2012.00124
Shah KU et al (2010) Voltage gated sodium channel blockers: potential treatment for neuropathic
pain. Curr Res Inform Pharm Sci 11:11–16
Shao N et al (2018) Familial hemiplegic migraine type 3 (FHM3) with an SCN1A Mutation in a
Chinese family: a case report. Front Neurol 9. https://doi.org/10.3389/fneur.2018.00976
Sokolov S, Scheuer T, Catterall WA (2005) Ion permeation through a voltage- sensitive gating pore
in brain sodium channels having voltage sensor mutations. Neuron 47:183–189. https://doi.org/
10.1016/j.neuron.2005.06.012
634 V. Tiwari et al.
Sokolov S, Scheuer T, Catterall WA (2007) Gating pore current in an inherited ion channelopathy.
Nature 446:76–78. https://doi.org/10.1038/nature05598
Tata A-G (2013) Effects of cysteine mutations in the S4-S5 linkers from domains D1 to D4 on the
fast inactivation of the voltage-gated sodium channel Nav1. 4. Universität Ulm. Medizinische
Fakultät, Ulm
Wang J et al (1992) Sequence and genomic structure of the human adult skeletal muscle sodium
channel α subunit gene on 17q. Biochem Biophys Res Commun 182:794–801. https://doi.org/
10.1016/0006-291X(92)91802-W
Watanabe K, Sato I, Kosaki T (1971) Familial hemiplegic migraine. No to Hattatsu. https://doi.org/
10.11251/ojjscn1969.3.14
Waxman SG, Kocsis JD, Black JA (2017) Type III sodium channel mRNA is expressed in
embryonic but not adult spinal sensory neurons, and is reexpressed following axotomy. J
Neurophysiol 72:466–470. https://doi.org/10.1152/jn.1994.72.1.466
Wood JN et al (2004) Voltage-gated sodium channels and pain pathways. J Neurobiol 61:55–71.
https://doi.org/10.1002/neu.20094
Yamagishi T et al (2001) Molecular architecture of the voltage-dependent Na channel: functional
evidence for alpha helices in the pore. J Gen Physiol 118:171–182. https://doi.org/10.1085/jgp.
118.2.171
Ye M et al (2018) Differential roles of Na V 1.2 and Na V 1.6 in regulating neuronal excitability at
febrile temperature and distinct contributions to febrile seizures. Sci Rep 8:753. https://doi.org/
10.1038/s41598-017-17344-8
Pharmacology of Potassium Channels
19
Satyendra Deka, Pobitra Borah, Ratnali Bania, Sanjib Das,
and Pran Kishore Deb
Abstract
Potassium channels constitute the largest and most ubiquitous and diverse ion
channel family. They regulate several physiological processes including electrical
signalling, hormone and neurotransmitter release, muscle contraction, cardiac func-
tion, cell proliferation and immune function, which are underscored by the associa-
tion of K+ channel mutations to numerous inherited diseases. Although many
challenges remain unsolved, the availability of diverse expression systems, molecu-
lar cloning and genetic linkage analysis has led to the upgradation of available
topological data, identification of disease-producing loci, and better understanding
of mutation-linked channelopathies. Moreover, in 2016, a new nomenclature
(KNa1.1 for KCa4.1, and KNa1.2 for KCa4.2) has been assigned and implemented
in the IUPHAR database. These advances along with high-throughput screening are
catalysing the discovery process of newer pharmacophores and modulators of K+
channels. This chapter aims to provide a basic understanding of K+ channels and
offers an updated overview on the progress and opportunities of pharmacological
approaches in the exploitation of these channels as therapeutic targets.
Keywords
Potassium channel · Ca2+- and Na+-activated K+ channels · Inwardly rectifying
K+ channels · Two P domain K+ channels · Voltage-gated K+ channels
Abbreviations
1-EBIO 1-Ethyl-2-benzimidazolinone
4-AP 4-Aminopyridine
AF Atrial Fibrillation
AgTx Agitoxins
ALS Amyotropic Lateral Sclerosis
BK Large conductance K+ channels
CHF Congestive Heart Failure
ChTx Charybdotoxin
CLL Chronic Lymphoid Leukaemia
CNS Central Nervous System
DCT Distal Convoluted Tubule
DRG Dorsal Root Ganglia
DTx Dendrotoxin
EA1 Episodic Ataxia type 1
EAG Ether-à-go-go
EGTA Ethylene Glycol Tetraacetic Acid
Elk Eag-Like K+ Channels
Erg Eag-Related Genes
GABA γ-Amino Butyric Acid
GAs General Anaesthetics
GIRK G-protein-activated Inward-Rectifier K+ channels
HERG Human ether-à-go-go
HMs Hypoglossal Motoneurons
HTS High-Throughput Screening
IbTx Iberiotoxin
IK Intermediate Conductance K+ channels
IUPHAR International Union of Basic and Clinical Pharmacology
K2P Two P domain K+ channels
KATP ATP-sensitive K+ channels
KCO Potassium Channel Opener
Kir Inwardly rectifying K+ channels
KT Kaliotoxin
mAb Monoclonal Antibodies
MAC Minimum Alveolar Concentration
MEM Memantine
MgT Margatoxin
MOR μ-Opioid Receptor
mRNA Messenger RNA
MTx Maurotoxin
NBP 3-N-Butylphthalide
NMDAR N-Methyl-D-aspartate receptor
NSAIDs Non-Steroidal Anti-Inflammatory Drugs
19 Pharmacology of Potassium Channels 637
NxTx Noxiustoxin
PHHI Persistent Hyperinsulinaemic Hypoglycaemia of Infancy
PIP2 Phosphatidylinositol-4,5-bisphosphate
PTX Pertussis toxin
RBCs Red Blood Cells
SCLC Small-Cell Lung Cancer cells
SeSAME Seizures, Sensorineural deafness, Ataxia, Mental retardation, Electro-
lyte imbalance
SK Small conductance K+ channels
SSRIs Selective Serotonin Reuptake Inhibitors
SU Sulfonylureas
SUR Sulfonylurea Receptors
T2-DM Type II Diabetes Mellitus
TCAs Tricyclic Antidepressants
TEA Tetraethylammonium
TM Transmembrane domains
TRAM-34 Triarylmethane-34
19.1 Introduction
Potassium channels form the largest and most diverse family among the ion channels
encoded by about 78 genes (D’amico et al. 2013). The first atomic structure of a
prokaryotic K+ channel, KcsA, was determined from bacterium Streptomyces
lividans (Kuang et al. 2015; Garcia et al. 2001; Luzhkov and Åqvist 2005; González
et al. 2012). After this discovery, considerable efforts have been made to interpret the
mechanism of the subtypes of the channel. The genomes of Drosophila and
Caenorhabditis elegans were found to contain 30–100 genes for K+ channels (Miller
2000). The subunit stoichiometry, topology, precise biophysical properties, and
modulation by the ligands and second messengers have been addressed to a great
extent for the K+ channels. These studies have been conducted by using high-
hroughput multiple assays, including T1+ flux assay, ligand binding assay,
voltage-sensitive dye-assay and 86Rb+ flux assay (Yu et al. 2016; Longman and
Hamilton 1992). Potassium channels switch the conformation between an open and
a closed state to conduct K+ ions down the electrochemical gradient of K+ across the
cell membrane (Mackinnon 2003). A typical K+ channel consists of a tetramer with
each monomer possessing one pore-forming (P) domain, which together comprises
the pore for conduction. The potassium channels contain a highly conserved seg-
ment of amino acid sequences known as K+ channel signature sequences (TVGYG)
(Kuang et al. 2015; Miller 2000). These sequences form the selectivity filter for K+
ions, which gives the channels more selectivity for K+ (at least 10,000 times) over
Na+ ions. The specificity is due to the multiple binding sites of the selectivity filter
that mimics a potassium ion hydrated shell. Moreover, the rate of conduction of these
channels is very high (107 ions channel1 s1) (Mackinnon 2003). The K+ channels
exist in three states, i.e. the channel is closed in the resting state, open in the activated
638 S. Deka et al.
state and remains non-conductive in the inactivated state. The gating is formed by
the bending of inner helix (intracellular) and the selectivity filter (extracellular)
(Kuang et al. 2015). The wide tissue distribution allows potassium channels to
influence various physiological functions, such as release of neurotransmitters and
hormones, regulation of fluid secretion, controlling of heart rate, smooth and cardiac
muscle contraction and clonal expansion of immune cells (Garcia et al. 1997; Garcia
and Kaczorowski 2005). The activation of K+ channels also regulates cellular
excitability by influencing the action potential waveform. In excitable cells, the
pharmacological activation of these channels reduces the excitability, albeit channel
inhibition leads to excitability of the cell.
Several genetically linked and acquired diseases have been known to be associated
with the alteration of the K+ channel. Disruption of the K+ channel gene underlies
various pathologies involving epilepsy, neurodegeneration, schizophrenia, episodic
ataxia, diabetes, deafness, cardiac arrhythmias, renal diseases, asthma and hyperten-
sion (Bergeron and Bingham 2012). This provides a basis to develop appropriate
pharmacological interventions by targeting these channels. Most interestingly, the
enthusiasm towards this arena was driven by the realization of the fact that the
sulfonylurea class of antidiabetic drugs and class-III antiarrhythmic drugs act via
regulating the K+ channels. Not only this, many K+ channel openers and blockers
also offer therapeutic opportunities in a wide range of areas, including vascular or
non-vascular muscle and immune, neuronal and cardiac systems.
Although continuous progress has been made in developing the underlying
molecular pharmacology of the K+ channels, still some of the subtypes remain
unexplored and in most of the cases existing tools are not sufficient for probing
these subfamilies in higher biological systems (Garcia and Kaczorowski 2016).
Therefore, emphasis has been laid on the discovery of K+ channel modulators in
the last few decades. The basic K+ channel modulators are mainly divided into two
types: peptide venom toxins and small molecules. The peptide toxins exert their
effect either by binding to the outer vestibule (e.g. toxins from snakes, sea anemones,
scorpions and cone snails) or by interacting with the voltage sensor of the channel
(e.g. hanatoxin obtained from spiders). However, the small molecules target the
gating hinge, inner pore or α/β subunit interface. The small molecules either activate
or block the channel. Depending on their selectivity towards target(s), they are also
classified as specific target and multi-target molecules (Tian et al. 2014).
According to the classification of IUPHAR, four structural types of K+ channels
are (a) Ca2+- and Na+-activated K+ channels, (b) inwardly rectifying K+ channels,
(c) two P domain K+ channels and (d) voltage-gated K+ channels (Alexander et al.
2017; Alexander et al. 2013). These individual classes are explained in detail in the
later sections. Moreover, expression of the channel subtypes and their modulators
are summarized in Table 19.1.
Table 19.1 Overview of potassium channels and their modulators
19
Modulators
Family Subfamily Members Gene Tissue expression Activators Inhibitors/Blockers References
Ca2+- and Large KCa1.1 KCNMA1 Ubiquitous; brain, NS004, NS1619, Paxilline, TEA, ChTx, Kaczmarek
Na+- conductance hair cells of cochlea, cromakalim, IbTx, slotoxin, EGTA, et al. (2017);
activated (BK) pancreatic β-cells, dehydrosoyasaponin-I, Ni+, aflatrem, Wei et al.
(KCa and skeletal and smooth niflumic acid, MCI-154, verruculogen, penitrem (2005); Coghlan
KNa) muscle, colon, Maxi-K diol, CGS-7181, A et al. (2001);
kidney NS-1608, Kaczorowski
BMS-20435211 et al. (1996);
KCa5.1 KCNU1 Testis, – Ba2+, quinine, quinidine Sanchez and
spermatocytes McManus
KNa1.1 KCNT1 Brain, kidney, testis, Bithionol, niclosamide, Bepridil, quinidine (1996);
hippocampus, loxapine Augustynek
Pharmacology of Potassium Channels
(continued)
Table 19.1 (continued)
640
Modulators
Family Subfamily Members Gene Tissue expression Activators Inhibitors/Blockers References
KCa2.3 KCNN3 Brain, lymphocytes, EBIO, NS309
skeletal muscle,
prostate, kidney,
vascular
endothelium, heart,
pancreas, colon,
liver, head, neck,
ovary
Intermediate KCa3.1 KCNN4 RBCs, liver, EBIO, NS309, SK-121, TRAM-34, ChTx, IbTx,
conductance pancreas, benzoxazoles maurotoxin,
(IK) lymphocytes, (zoxazolamine and clotrimazole,
placenta, prostate, chlorzoxazone), isatin nitrendipine, oxime,
colon, lung, colon, derivatives malonate
vascular
endothelium
Inwardly Classical or Kir2.1 KCNJ2 Heart, brain, kidney, PIP2 Spermine, spermidine, González et al.
rectifying strong lung, placenta, putrescine, intracellular (2012); Garcia
(Kir) inward skeletal and smooth Mg2+, Ba2+, Cs+, Rb+, and
rectifier muscle, memantine Kaczorowski
macrophage (2005);
Kir2.2 KCNJ12 Cerebellum, Intracellular Mg2+, Ba2+, Alexander et al.
forebrain, heart, Cs+ (2017); Shieh
kidney, skeletal et al. (2000);
muscle Yamashita et al.
Kir2.3 KCNJ4 Brain, reactive Intracellular alkalization Intracellular Mg2+, Ba2+, (1996); Ishihara
astrocyte, kidney, (pH 6.76), PIP2, Cs+, spermine, et al. (1996);
heart, smooth arachidonic acid, tenidap spermidine, putrescine Alagem et al.
muscle (2001); Hughes
S. Deka et al.
19
G-protein Kir2.4 KCNJ14 Brain, retina, Extracellular alkalization Intracellular Mg2+, Ba2+, et al. (2000);
activated neuronal cell in Cs+ Camerino et al.
(GIRK) heart (2007)
brain, heart
(continued)
Table 19.1 (continued)
642
Modulators
Family Subfamily Members Gene Tissue expression Activators Inhibitors/Blockers References
K+-transport Kir1.1 KCNJ1 Kidney, pancreatic VU590, VU591 Tertiapin-Q,
islets, skeletal δ-dendrotoxin, Ba2+, Cs+
muscle, pancreas,
spleen, brain, liver,
heart
Kir4.1 KCNJ10 Glia, retina, ear, ATP, PIP2 Ba2+, Cs+, imipramine,
kidney desipramine,
nortryptyline,
amitriptyline
Kir4.2 KCNJ15 Brain, kidney, lung, – Ba2+, Cs+
pancreas, liver,
testis
Kir5.1 KCNJ16 Brain, kidney, – Ba2+, Cs+
thyroid, spleen,
liver, testis, retina
Kir7.1 KCNJ13 Cerebellum, – Ba2+, Cs+
hippocampus,
kidney, thyroid, GI,
stomach, small
intestine, prostate,
testis, lung, retina
Two P TWIK K2P1.1 KCNK1 Brain, heart, kidney, – Garcia et al.
domain or lung, liver, placenta (1997); Gada
(K2P) TWIK-1 and Plant
S. Deka et al.
19
K2P6.1 KCNK6 Pancreas, heart, Arachidonic acid Ba2+, quinidine, volatile (2019); Enyedi
or placenta, lung, anaesthetics and Czirják
TWIK-2 stomach, eyes, (2010);
embryo Olschewski
K2P7.1 KCNK7 Brain, spinal cord, et al. (2017);
retina Vivier et al.
TREK K2P2.1 KCNK2 Brain, lung, heart Arachidonic acid, Ba2+, PKA, PKC, (2015); Lesage
or Chloroform, halothane, sipatrigine, fluoxetine, (2003); Wright
TREK-1 isoflurane, unsaturated chlorpromazine, et al. (2017);
fatty acid, haloperidol, loxapine, Tian et al.
lysophospholipids, pimozide, fluphenazine, (2019); Hayashi
fenamates, flufenamic mexiletine, propafenone, and Novak
acid, GI-530139 amlodipine, nifedipine (2013);
Staudacher et al.
Pharmacology of Potassium Channels
Modulators
Family Subfamily Members Gene Tissue expression Activators Inhibitors/Blockers References
K2P9.1 KCNK9 Brain Halothane External pH (6.5),
or ruthenium red,
TASK-3 anandamide, R-(+)-
methanandamide
KCNK15 Brain, kidney, heart, – –
lung, pancreas,
liver, placenta,
thyroid, adrenal
gland, salivary
K2P15.1 gland
or
TASK-5
TALK K2P5.1 KCNK5 Brain, kidney, liver, Halothane, volatile External pH (6.5), LAs
or pancreas, placenta, anaesthetics (lidocaine, bupivacaine,
TASK-2 small intestine clofilium)
K2P16.1 KCNK16 Heart, liver, lung, Isoflurane, NO, ROS External pH, Ba2+,
or pancreas, placenta quinidine, chloroform
TALK-1
K2P17.1 KCNK17 Heart, lung, liver, NO, ROS, quinidine, External pH, Ba2+,
or placenta, pancreas propafenone, mexiletine, chloroform, sotalol,
TALK-2 metoprolol, propranolol verapamil, amiodarone,
ranolazine
THIK K2P12.1 KCNK12 Brain, liver, lung, – Propafenone, mexiletine,
or heart, kidney, colon, propranolol, lidocaine
THIK-1 pancreas, spleen,
ovary, placenta,
prostate, thymus,
small intestine
S. Deka et al.
19
(continued)
645
Table 19.1 (continued)
646
Modulators
Family Subfamily Members Gene Tissue expression Activators Inhibitors/Blockers References
Kv1.4 KCNA4 Corpus striatum, – Fampridine, UK78282, Vacher et al.
hippocampus, riluzole, quinidine, (2007);
olfactory bulb, nicardipine Camerino et al.
pancreatic islets, (2007);
heart, skeletal and Alexander et al.
smooth muscle, (2013)
lung carcinoid
(continued)
Table 19.1 (continued)
648
Modulators
Family Subfamily Members Gene Tissue expression Activators Inhibitors/Blockers References
fast-GABAergic 3-isobutyl-1-
interneurons, methylxanthine
Schwann cells,
mesenteric artery
Kv3.3 KCNC3 Brainstem, – 4-AP, TEA
forebrain, Purkinje
cells, cerebellum,
motoneurons, lens,
corneal epithelium
Kv3.4 KCNC4 Brainstem, – 4-AP, TEA, sea anemone
hippocampus, toxin BDS-I
skeletal muscle,
parathyroid,
prostate, pancreatic
acinar cells
Kv4 Kv4.1 KCND1 Heart, liver, kidney, – Fampridine
pancreas, thyroid
gland, lung,
stomach, testis,
pulmonary artery
Kv4.2 KCND2 Cerebellum, – Hanatoxin,
thalamus, basal heteropodatoxins
ganglia,
hippocampus,
forebrain, cochlear
nucleus, rodent
heart
S. Deka et al.
19
(continued)
Table 19.1 (continued)
650
Modulators
Family Subfamily Members Gene Tissue expression Activators Inhibitors/Blockers References
cerebellum,
brainstem, cerebral
cortex, lung, testis,
breast, eye,
placenta, small
intestine, DRG,
sympathetic ganglia
Kv7.3 KCNQ3 Hippocampus, Retigabine Linopirdine, 4-AP,
cerebral cortex, clofilium, CTX, E4031
cerebellum,
brainstem,
thalamus, DRG,
sympathetic
ganglia, colon, eye,
head, neck, retina
Kv7.4 KCNQ4 VSM, outer hair Retigabine XE991, linopirdine,
cells of ear and TEA, bepridil
cochlea, placenta
Kv7.5 KCNQ5 Cerebral cortex, Retigabine, BMS204352 XE991, linopirdine
thalamus,
hippocampus,
skeletal muscle,
VSM, sympathetic
ganglia
Kv8 Kv8.1 KCNV1 Kidney, brain – –
Kv8.2 KCNV2 Lung, kidney, liver, – –
thymus, pancreas,
S. Deka et al.
19
Modulators
Family Subfamily Members Gene Tissue expression Activators Inhibitors/Blockers References
uterus, lactotrophs,
rat pituitary
Kv11.3 KCNH7 Brain, sympathetic – Sertindole, pimozide
ganglia, lactotrophs,
CA pyramidal
neurons, rat
pituitary
Kv12 Kv12.1 KCNH8 Brain, sympathetic – Ba2+
ganglia, lung,
uterus, colon, testis
Kv12.2 KCNH3 Infant brain, – Ba2+
amygdala,
hippocampus, eye
(retinoblastoma),
lung (small-cell
carcinoma)
Kv12.3 KCNH4 Telencephalon, – Ba2+
lung, oesophagus,
pituitary,
cerebellum,
neuroblastoma,
primary B-cell
neoplasia,
oligodendroglioma
S. Deka et al.
19 Pharmacology of Potassium Channels 653
Studies demonstrated that chelation of Ca2+ reduces the K+ efflux in RBCs, while K+
conductance increases after intracellular injection of Ca2+ into neurons (Kaczmarek
et al. 2017). Based on these observations, the first gene from a Drosophila mutant
(slowpoke or slo) encoding a Ca2+-activated K+ channel was cloned and identified
(Kaczmarek et al. 2017; Atkinson et al. 1991). This channel was termed BK or
MaxiK (later named KCa1.1 or Slo1) because of its large conductance capacity and
sensitivity towards both Ca2+ and transmembrane voltage (Contreras et al. 2013;
Greenwood and Leblanc 2007). Screening of cDNA libraries for similar sequences
of a K+-selective pore subsequently led to the discovery of different genes encoding
for other Ca2+-activated K+ channel subtypes. The next two identified classes,
i.e. KCa2 or SK (small conductance) family (KCa2.1 or SK1, KCa2.2 or SK2, and
KCa2.3 or SK3) and KCa3 or IK (intermediate conductance) family (KCa3.1 or SK4),
are insensitive to the membrane voltage (Adelman et al. 2012). Based on structural
resemblance, three other genes were identified and named KCa4.1 (Slack or Slo2.2),
KCa4.2 (Slick or Slo2.1) and KCa5.1 (SLO3).
Later it was observed that KCa4.1 (Slack) and KCa4.2 (Slick) are not only
activated by the intracellular Ca2+ but also regulated by the Na+ and Cl concentra-
tion in the cytoplasm (Kaczmarek 2013). Based on this, a new nomenclature for the
two channels (KNa1.1 for Slack or former KCa4.1, and KNa1.2 for Slick or former
KCa4.2) was proposed by Kaczmarek et al. (2017), and later it was accepted and
implemented in the IUPHAR database (Alexander et al. 2017; Kaczmarek et al.
2017).
Cryo-electron microscopy and X-ray crystallography revealed that KCa1.1
consists of 7-TM domains distinguishing it from canonical 6-TM K+ channels. It
has an additional S0 domain preceding S1 along with the common S1–S6 domain
sequence with N-terminal lying outside rather than inside of the cell (as shown
in Fig. 19.1) (Meera et al. 1997). The RCK1 and RCK2 domains in the cytoplasmic
C-terminal act as binding sites for Ca2+, and the “gating ring” is formed together by
the eight RCK domains of each tetrameric KCa1.1 channel (Wu et al. 2010; Wei et al.
2005). Unlike the KCa1.1 channel, KCa2 channels contain six α-helical TM (S1–S6)
with a K+-selective pore sequence linking S5 and S6 (Kaczmarek et al. 2017). These
channels are voltage independent, and conduction through the channels occurs only
after Ca2+–calmodulin complexation (Xia et al. 1998). Similarly, KCa3.1 closely
resembles the KCa2 family; however, unitary conductance of this channel is greater
compared to KCa2. Therefore, it is also named intermediate conductance for the K+
or IK channel. The sensitivity of KCa3.1 is also determined by the association of Ca2+ to
calmodulin. KNa1.1 and KNa1.2 differ from KCa1.1 in transmembrane topology as
they lack the S0-TM. Though these are voltage-sensitive channels, they lack the
repeated motif of amino acids in S4 segments like KCa1.1. The two RCK domains
form the gating ring of the pore similar to that of KCa1.1. The KCa5.1 has a similar
topology as KCa1.1 (Kaczmarek et al. 2017). KCa5.1 is expressed mainly in sperm
cells and activated primarily by voltage and internal alkalinization as in the case of
sperm capacitation (Navarro et al. 2007; Zeng et al. 2015).
654
+
+ +
+
+ +
S0 1 2 3 4 5 6
1 2 3 4 5 6 1 2 3 4 5 6
+
+ +
+
+ +
CaM
RCK2
RCK2
(a) (b) (c)
+++ +++
+
+
1 2 1 2 3 4 1 2 3 4 5 6
+
+
Fig. 19.1 Schematic diagram of the transmembrane topology of different potassium channels: (a) Large conductance KCa1.1 and KCa5.1 channels with
additional S0 domain in the structure and Ca2+-binding sites RCK1 and RCK2, (b) small and intermediate conductance KCa2.1, KCa2.2, KCa2.3 and KCa3.1
channels with calmodulin as the Ca2+-binding site, (c) large conductance KNa1.1 and KNa1.2 (formerly known as KCa4.1 and KCa4.2) channels with RCK1 and
RCK2 subunits (without S0 domain), (d) inwardly rectifying (Kir) channels or a typical 2-TM domain family, (e) two P domain (K2P) or 4-TM potassium
channels with two P-loops forming a tandem and (f) voltage-gated K+ channels (Kv) with 6-TM domain comprising homomeric and heteromeric tetramers
S. Deka et al.
19 Pharmacology of Potassium Channels 655
(oestrogen receptor antagonist) at its therapeutic concentration may explain their role
in tamoxifen-induced QT prolongation and arrhythmia.
The scorpion toxins iberiotoxin (IbTx), charybdotoxin (ChTx) and slotoxin are
specific blockers of KCa1.1 with higher selectivity and potency (Garcia-Valdes et al.
2001). These channels are also blocked by tetraethylammonium (TEA), Ni+ and
ethylene glycol tetraacetic acid (EGTA) (Dogan et al. 2019). Non-selective vascular
BK blockers include gallopamil and verapamil. BK channels are also inhibited by
ketamine (dissociative anaesthetic) and clotrimazole (antifungal) (also refer to
Table 19.1) (Kaczmarek et al. 2017).
No specific pharmacological agents are known to selectively act on KNa1.1,
KNa1.2 and KCa5.1. Drugs such as bithionol, niclosamide and loxapine can
pharmacologically activate these channels. Loxapine is more selective towards
KNa1.1 unlike bithionol, which also activates KCa1.1. Quinidine is known to ame-
liorate the symptoms of malignant migrating partial seizures in infancy by blocking
KNa1.1 currents (Bearden et al. 2014; Yang et al. 2006; De Los Angeles Tejada et al.
2012). The mechanism of quinidine-mediated amelioration of these symptoms is not
very clear as selectivity of quinidine is not limited to KNa1.1 channels. Various
non-specific KNa1.1 blockers include Ba2+, clofilium and bepredil (Bhattacharjee
et al. 2003).
Similarly, KNa1.2 is less explored and reportedly activated by the fenamate class
of NSAIDs like niflumic acid, with low potency. These agents are known to
uncouple the channels from Na+ or transmembrane voltage-regulated modulation
causing greater conductance of current. As they affect other channels including
KCa1.1, this action is non-specific in nature. Other pharmacological blockers of
these channels include clofilium, isoflurane and quinidine (Bhattacharjee et al.
2003; Berg et al. 2007).
The selective pharmacological agents of KCa5.1 are not known yet. However, this
channel is blocked by Ba2+, quinine and quinidine up to some extent (Sánchez-
Carranza et al. 2015; Tang et al. 2010).
Sir Bernard Katz first described the membrane conductance attributed to Kir
channels. He discovered that in frog skeletal muscle fibres, K+ ions move more
readily into the inside compared to the outside (called inward rectification) (Doupnik
2017). These channels were so named because of this characteristic degree of inward
rectification, i.e. prominent asymmetrical conduction of potassium in the inward
direction compared to the outward current (Coghlan et al. 2001). The inward
rectification is observed due to the blockade of depolarized potential (outward
current) by intracellular Mg2+ ions, natural polyamines, putrecine, spermidine and
spermine (Kurata et al. 2004; Köhling and Wolfart 2016; Jiménez-Vargas et al.
2017; Nichols and Lopatin 1997).
X-ray crystallography has detailed the 3D structure of Kir channels to the atomic
level. Structurally, Kir channels are the simplest among the K+ channel families, with
four subunits formed of 2-TM domains separated by a segment of pore-forming
(P) elements (as shown in Fig. 19.1) (Humphries and Dart 2015). The transmem-
brane domain regulates the gating and ion selectivity, whereas the cytoplasmic
domain is involved in the control of gating by G-proteins, Na+ ions and nucleotides.
658 S. Deka et al.
The Kir channels are widely distributed in neurons, glial cells, cardiac myocytes,
blood cells, osteoclasts, endothelium, epithelium and oocytes exhibiting distinct
roles in both normal and pathophysiological conditions. Alteration in the functions
of these channels may lead to several genetically linked and acquired diseases.
Genetic studies have linked many rare human diseases with Kir channel gene
mutations. For example, recessive loss-of-function mutations in the KCNJ1 gene
or Kir1.1 cause type II Bartter syndrome (Doupnik 2017; Dworakowska and Dolowy
2000), and loss-of-function mutations in Kir2.1 are associated with Andersen syn-
drome (LQT7) (Hibino et al. 2010; Giudicessi and Ackerman 2012). Acquired
pathological conditions such as electric remodelling associated with atrial fibrillation
(AF) can occur due to upregulation of Kir2.1 and Kir3.1/3.4 channel activity. KATP
channels are implicated in glucose metabolism and contractility of heart.
Generalized seizures and SeSAME syndrome are associated with Kir4.1
channelopathy (Bhave et al. 2010; Seifert et al. 2018). Permanent neonatal diabetes
occurs due to gain-of-function mutation in Kir6.2/SUR1. Moreover, new therapeutic
approaches, such as designing of K+ sparing diuretics for CHF and HT by targeting
renal Kir1.1 channels and treating AF by selectively inhibiting atrial Kir3 channels,
have spurred research interest towards this family.
At physiological voltage, generation of PIP2 by ATP-dependent kinases activates
Kir channels (Huang et al. 1998). Ba2+ and Cs+ (classical blockers) are known for
19 Pharmacology of Potassium Channels 659
effective blocking of majority of the Kir channels and are used to explore their
physiological roles (González et al. 2012). This inhibition is more prominent when
the membrane is hyperpolarized. Despite limited number of Kir channel blockers,
physiological and pharmacological assays have explored compounds with selectiv-
ity towards particular types of Kir channels. These compounds are discussed under
specific subtypes.
the three compounds (Robertson and Steinberg 1990; Edwards and Weston 1995).
The pharmacological properties are known to produce, only when there is
co-expression of Kir6.2 with an appropriate SUR subtype leading to KATP conduc-
tance (Lawson 2000; Quast 1992). The side effects of SUs are also related to their
ability to cross-react with other KATP subtypes. Bimakalin (a selective KCO)
exhibited a dose-dependent vasodilation but failed to show anti-ischemic benefits
in patients with coronary artery disease (CAD) during exercise-induced angina
pectoris (Chan et al. 2008). Levosimendan, a KATP channel opener, showed signifi-
cant reduction in pulmonary capillary wedge pressure in severe low-output HF
patients following cardiac surgery and peripartum cardiomyopathy. Levosimendan
has entered into the clinical studies for heart failure, amyotrophic lateral sclerosis,
ventricular dysfunction, myocardial infarction, hip fracture and cardiorenal syn-
drome (Rubaiy 2016). Another new antihypertensive drug, iptakalim, synthesized
by Thadweik Academy of Medicine, China, also activates KATP channels in the
endothelium of resistance blood vessels (Wang et al. 2015; Duan et al. 2011).
Iptakalim (KCO) as well as fluoxetine (classical antidepressant) showed alleviation
of chronic mild stress depressive behaviour in wild-type mice. However, these
symptoms are partially ameliorated in Kir6.2/ mice (Fan et al. 2016).
5-Hydroxydecanoic acid (5-HD) is a selective mitochondrial and plasma mem-
brane KATP channel blocker. It is highly used in the study of the physiological role of
mito-KATP channels. Non-sulfonylurea KATP channel blockers such as meglitinide,
nateglinide, repaglinide and mitiglinide are also used to treat T2-DM. These drugs
act by inhibiting pancreatic β-cell KATP channels, i.e. blocking Kir6.2/SUR1
channels. Many derivatives of P1075, a cyanoguanidine K+ opener, have been
known to antagonize the vascular KCOs. These new-generation drugs called PNU
compounds include PNU-37883A, PNU-89692, PNU-97025E and PNU-99963
(Rubaiy 2016; Chowdhury et al. 2017).
pore of Kir4.1 channels. No effective blockers of Kir5.1 are reported till date. While
Kir7.1 remains unusually insensitive to the typical blocking property of Ba+ and Cs+,
interestingly, no other activators or blockers are known for this channel.
The first potassium channel containing two P domain in tandem was the TOK-1
channel discovered from the yeast Saccharomyces cerevisiae (Goldstein et al. 2001;
Ketchum et al. 1995). These channels are referred to as “twin pore” or “tandem pore”
as they possess nonconventional topology, i.e. dimers of dimers: two subunits
forming a mature channel containing two non-identical pore-forming P domains
(P1 and P2) which are arranged in tandem (as shown in Fig. 19.1) (Tian et al. 2014;
Humphries and Dart 2015; Schneider et al. 2014). Most of the K2P channels conduct
voltage-independent outward current as they lack the voltage-sensing S4-TM
domain (Coghlan et al. 2001; Goldstein et al. 2001). These channels also act as the
classical background “leak” K+ channels or open rectifiers and maintain negative
resting membrane potential (Köhling and Wolfart 2016; Piechotta et al. 2011).
Fifteen mammalian KCNK genes encode K2P channels. These channels were
distributed among six clades on the basis of sequence homology and pharmacologi-
cal characteristics. These are as follows: (1) two-pore domain weak inward-
rectifying K+ channels or TWIK (K2P1 or TWIK-1, K2P6 or TWIK-2, and K2P7)
are weak inward rectifiers sensitive to pH; (2) TWIK-related K+ channels or TREK
(K2P2 or TREK-1, K2P4 or TRAAK, and K2P10 or TREK-2) are mechano-gated
channels regulated by several stimuli – pH, stretch, osmolarity, temperature,
neuroprotective agents and volatile anaesthetics; (3) TWIK-related acid-sensitive
K+ channels or TASK (K2P3 or TASK-1, K2P9 or TASK-3, K2P15 or TASK-5) are
inhibited by extracellular acidification and hypoxia; (4) TWIK-related alkaline
pH-activated K+ channels or TALK (K2P5 or TASK-2, K2P16 or TALK-1, and
K2P17 or TALK-2) are sensitive to alkaline pH and oxygen concentration; (5) tandem
pore domain halothane-inhibited K+ channels or THIK (K2P12 or THIK-2, K2P13 or
THIK-1) are sensitive to halothane and arachidonic acid; (6) TWIK-related spinal
cord K+ channels or TRESK (K2P18) are sensitive to free acids, protons, heat,
anaesthetics and increased membrane tension (Jiménez-Vargas et al. 2017;
Feliciangeli et al. 2015; Renigunta et al. 2015; Talley et al. 2003; Lesage and
Lazdunski 2000).
Given their critical physiological roles, K2P channels represent a major target for the
modulation of the physiological changes and pathological states. For example, K2P1
and K2P2 channels play an important role in the regulation of atrial size and heart rate
and are, therefore, a putative target for antiarrhythmic drugs. Similarly, K2P18
channels are good candidates for pain management as the pain signalling pathway
19 Pharmacology of Potassium Channels 663
is strongly controlled by the K2P18 channels. Moreover, K2P channels are implicated
in arrhythmia (K2P1), depression (K2P2), atrial fibrillation (K2P3), migraine (K2P18),
cancer (K2P1, K2P5, K2P9) and inflammation (K2P12/13) as well as in the neuro-
pathic pain (K2P18) (also refer to Table 19.1) (Feliciangeli et al. 2015; Gada and
Plant 2019; Li and Toyoda 2015).
Considering the lead optimization and drug discovery process, several practical
limitations were observed. The unique structure of the K2P family restricted the easy
extrapolation from the available data of other K+ channel subfamilies. The lack of a
structural basis of the K2P channels, until the explanation of the crystal structure of
K2P1 and K2P4 in 2012, had stymied the development of pharmacophores of these
channels (Gada and Plant 2019). Moreover, the electrophysiological characterization
for understanding the biological significance of K2P channels has been impeded by
the poor or absent heterologous expression in Xenopus oocytes or COS-7 cells
(Enyedi and CzirjáK 2010). The ubiquitous distribution of many subunits also limits
the therapeutic modulation of specific K2P channels. To develop any pharmacologi-
cal modulator of these subunits, the researchers must overcome these barriers with
available HTS techniques and necessary measures. Nevertheless, the numbers of
small molecules and drugs interacting with the K2P channels are steadily increasing
over time. These channels are weakly sensitive or insensitive to the classical K+
channel blockers including 4-AP, TEA, Cs+ and Ba+ (Dogan et al. 2019). Some
other potent modulators have already been identified and are discussed in the
succeeding section (Tian et al. 2014; Es-Salah-Lamoureux et al. 2010; Kasap and
Dwyer 2018; Gada and Plant 2019).
Sipatrigine, a neuronal Na+ and Ca2+ channel inhibitor, reversibly inhibits both
TREK-1 and TRAAK in a dose-dependent manner. This effect in combination with
glutamate inhibition makes this drug a choice for treatment of depression (Lesage
2003; Meadows et al. 2001; Tsai 2008). 3-N-Butylphthalide (NBP) extracted from
celery seed also reversibly inhibits TREK-1 current. This neuroprotective agent is
used in China for treatment of ischemic stroke (Ji et al. 2011). SSRIs such as
fluoxetine reversibly block TREK-1 and related TASK-3 channels in a voltage-
independent manner. Similarly, norfluoxetine, active metabolite of fluoxetine, is a
more potent blocker of TREK-1 current (Kennard et al. 2005). In addition to SSRIs,
several other antipsychotics such as chlorpromazine, loxapine, haloperidol,
pimozide and fluphenazine also exhibited dose-dependent and reversible inhibition
of both TREK-1 as well as TREK-2 but not TRAAK channels (Thümmler et al.
2007). Other pharmacological classes of drug inhibiting TREK-1 current include
antihypertensive dihydropyridines (e.g. amlodipine and nifedipine), calcium
antagonists (e.g. flunarizine) and antiarrhythmic agents (e.g. mexiletine and
propafenone) (also refer to Table 19.1) (Vivier et al. 2015).
Among volatile GAs, chloroform is the potent activator of TASK-2 (Patel and
HonorÉ 2001; Gray et al. 2000; Yost 2000). Clofilium inhibits TASK-2, while
drugs like quinidine, bupivacaine and lidocaine are strong inhibitors of TASK-
2 and weak inhibitors of TALK-1 (Hayashi and Novak 2013; Girard et al. 2001;
Gutman et al. 2005). Whole-cell patch clamp and two-electrode voltage clamp
studies demonstrated the sensitivity of antiarrhythmic drugs to TALK-2 channels
in the Xenopus oocytes expression system. Significant activation was exhibited by
quinidine, propafenone, mexiletine, metoprolol and propranolol, while drugs such as
sotalol, verapamil, amiodarone and ranolazine inhibit the TALK-2 current (also refer
to Table 19.1). Human TALK-2 channels were reported to be sensitive towards
multiple antiarrhythmic drugs (Staudacher et al. 2018a). Activation of the THIK-1
channel by arachidonic acid and inhibition by halothane allow this channel to be
easily distinguished from TREK, which is activated by halothane (Kim 2005).
Therapeutic modulation of THIK-1 channels can make them a suitable target of
antiarrhythmic drugs. Drugs such as propafenone, mexiletine, propranolol and
lidocaine are known to inhibit THIK-1 channels (Staudacher et al. 2018b).
TRESK channels are activated by volatile anaesthetics including isoflurane,
desflurane, sevoflurane and halothane within the clinically used concentrations.
Cloxyquin activates the TRESK channel independent of Ca2+/calcineurin associa-
tion and is hence used for examining TRESK conductions in native cells (Lengyel
et al. 2017). Additionally, TRESK (K2P18) is insensitive to several classical K+
channel blockers, such as 4-AP, apamin and CsCl, and KATP channel blockers, like
tolazamide and glipizide (also refer to Table 19.1). Arachidonic acid, quinine and
quinidine are potent blockers of this channel, while extracellular Ba2+-mediated
inhibition is observed at higher concentrations. Amide local anaesthetics such as
bupivacaine, tetracaine, rupivacaine, chlorprocaine, mepivacaine and lidocaine are
reported to influence mouse and human TRESK. About 240 substances were tested
on mouse TRESK. Among these, zinc, mercuric ions and mibefradil (non-specific to
K2P channels) were found to efficiently inhibit the current. Antidepressants, fluoxe-
tine and sipatrigine also block the TRESK current along with the TREK family
members, while lomotrigin (an anticonvulsant) showed selectivity towards only
TRESK (10-5M). Loratadine (an antihistaminic) also exhibited TRESK inhibition
with a micromolar IC50 (Enyedi and CzirjáK 2015).
The first ever cloned gene from Drosophila was the voltage-gated Shaker channel
followed by the phenotyping of the other potassium channel subtypes in insects, rats
and mammals including humans (Gutman et al. 2005). The Drosophila Shaker
channel acts as a prototype of all voltage-gated channels, consisting of a tetramer
of homologous α-subunits, each comprising 6-TM segments and a circumferentially
arranged membrane re-entering P-loop (Yellen 2002; Jensen et al. 2012). In brief,
the four S1–S4 segments act as voltage sensors, which contain arginine (positively
charged) in the S4 segment, while the K+ conduction pore is formed by the four
666 S. Deka et al.
S5-P-S6 sequences. The gating of the pore is regulated by pulling the S4-S5 linker
(Kuang et al. 2015; Pongs 1999; Sokolova et al. 2001; Kim and Nimigean 2016;
Wulff et al. 2009; Jackson 2017). Both the amino and carboxy terminals are found to
lie on the intracellular side of the membrane (as shown in Fig. 19.1). Membrane
depolarization activates majority of the Kv channels, whereas hyperpolarization
leads to closing of the channel. The Kv channels are encoded by about 40 genes
constituting the largest K+ family, which is categorized under 12 subfamilies. This
wide diversification of the channel arises from several factors, such as
heteromultimerization, modifier subunits, alternate mRNA splicing, post-
translational modification and association of other proteins like calmodulin and
minK (Gutman et al. 2005). The voltage-gated channels are mainly classified as
follows: Kv1 (Kv1.1–1.8), Kv2 (Kv2.1–2.2), Kv3 (3.1–3.4), Kv4 (Kv4.1–4.3), Kv5
(Kv5.1), Kv6 (Kv6.1–6.4), Kv7 (Kv7.1–7.5), Kv8 (Kv8.1–8.2), Kv9 (Kv9.1–9.3),
Kv10 (Kv10.1–10.2), Kv11 (Kv11.1–11.3) and Kv12 (Kv12.1–12.3) subfamilies.
The Kv channels are often expressed together with voltage-gated sodium or calcium
channels as they regulate the action potential firing. As the Kv current controls the
excitability of the cell, the pharmacological modulation of these channels may
regulate the cellular excitability. Therefore, Kv channels play a significant role in
Ca2+ signalling, proliferation, volume regulation and secretion as well as migration
(Wulff et al. 2009). Not only this, the Kv channel is also found to be involved in the
regulation of cellular activation in lymphocyte and cancer cells; consequently, Kv
blockers have showed effectiveness in inhibition of proliferation and cellular activa-
tion (Chandy et al. 2004; Pardo et al. 1999; Comes et al. 2013). Overall, mutations or
any defects in Kv channel genes are associated with a number of diseases, ranging
from autoimmune diseases to cancer to cardiovascular, neurological and metabolic
disorders. For example, episodic ataxia type 1, or EA1, is associated with loss-of-
function mutations of the KCNA1 gene (Kv1.1) (Maljevic and Lerche 2013;
Lehmann-Horn et al. 2003), whereas loss-of-function mutations of Kv1.1/ Kv1.2
lead to neuromyotonia associated with small-cell lung cancer (SCLC) cells and
limbic encephalitis (Camerino et al. 2007). Kv channels are also implicated in
neuronal apoptosis (Kv1.1, Kv1.3), ischemic cell death (Kv1.5), epilepsy (Kv1.1,
Kv1.2, Kv1.4, Kv3.2, Kv4.2, Kv4.3), Alzheimer’s disease (Kv3.4, Kv4.2), tinnitus
(Kv7.1– Kv7.5) pain (Kv7.1–Kv7.5) and neuropsychiatric disorders (Kv7.1–Kv7.5)
(also refer to Table 19.1) (Tsantoulas 2015; Shah and Aizenman 2014; Langguth
et al. 2016).
The pharmacological modulation of Kv channels can be effective in the therapeu-
tic management of various human channelopathies, which is further underlined by
the transgenic mice models. Metal ions, peptide toxins and small molecules are well
known to regulate Kv current conduction by either external or internal binding to the
conducting pore or by altering the channel gating mechanism (Wulff and Zhorov
19 Pharmacology of Potassium Channels 667
from sea anemone (e.g. BgK, ShK and AsKS) and spider venom (e.g. hanatoxin
1 and hanatoxin 2) are also well-known blockers of the Kv channels (Garcia et al.
1997). A natural triterpene, correolide, is a potent blocker of Kv1.3 but showed
affinity for other members of the Kv1 family (Felix et al. 1999). Pharmacological
agents such as naltrexone, sulfamidbenzamidoindane, UK-78282, WIN-17317-3
and CP 339818 have been extensively studied for their inhibitory effect on Kv1.3
channels (Coghlan et al. 2001; Hill et al. 1995).
Fairly mild acidosis (pH ¼ 6.5) can inhibit the Kv1.4 current due to the slow
recovery of N-type inactivation, which is a useful diagnostic characteristic of this
subtype (Claydon et al. 2000). Kv1.4 channels are blocked by 4-AP (0.7–13 mM) but
exhibit resistance to external TEA, DTx or similar toxins. Few small molecules are
known to inhibit the Kv1.4 current, including UK78282, riluzole, quinidine and
nicardipine (also refer to Table 19.1) (Gutman et al. 2005; Kuzmenkov et al. 2015).
Abundant distribution of the Kv1.5 channel makes it undoubtedly one of the most
important channels. They are insensitive to TEA while sensitive to 4-AP even at
relatively lower concentrations. Channel inhibition is exhibited by several therapeu-
tic classes including calcium channel blockers (e.g. verapamil and nifedipine),
antiarrythmic drugs (e.g. quinidine, clofilium and propafenone), antibiotics
(e.g. erythromycin) and antihistaminics (e.g. loratidine, terfenadine and ebastine)
(also refer to Table 19.1) (Zhang et al. 1997; Grissmer et al. 1994; Malayev et al.
1995; Gutman et al. 2005; Gutman et al. 2003).
Kv1.6 is sensitive to both TEA and 4-AP. Venoms like ChTx, MgTx, hongotoxin
and tumulustoxin were identified as the potent blockers of these channels. Kv1.7
channels are insensitive towards external TEA but blocked by 4-AP, nifedipine,
amiodarone, quinidine, flecainide and tedisamil, whereas reported blockers of Kv1.8
include Ba2+, ChTX, 4-AP, TEA, ketoconazole, pimozide and verapamil (Gutman
et al. 2005; Kuzmenkov et al. 2015; Gutman et al. 2003).
inactivation (Rudy and Mcbain 2001). Kv3 manifests extraordinarily high sensitivity
towards 4-AP and TEA because of the presence of the tyrosine residue at the
carboxyl terminal of P-loop. Drugs like cromakalim, diltiazem, nifedipine, flecainide
and resiniferatoxin also inhibit Kv3.1 channels. The Kv3.2 current is blocked by
8-bromo-cGMP, verapamil, D-NONOate and 3-isobutyl-1-methylxanthine. Kv3.3 is
reported to show variable inactivation kinetics in contrast to other Kv3 family
members. The subunits of Kv3.3 are sensitive to hypoxia and hence implicated in
oxygen sensing in pulmonary vasculature (Guo et al. 2008). Blood-depressing
substance (BDS) from sea anemone, Anemonia sulcata, has two isoforms, BDS I
and BDS II, which are selective and effective blockers of Kv3.4 (also refer
to Table 19.1). Although the specificity of BDS I towards Kv3.4 has been
questioned, these toxins at higher concentrations were earlier employed as selective
tools for Kv3.4 (Riazanski et al. 2001; Yeung et al. 2005).
Kv7.2 channels are resistant to 4-AP (2 mM) but are effectively blocked by TEA as
they possess the tyrosine residue in P-loop. Kv7.2 channels are also blocked by
XE991 and L-735821. In contrast, Kv7.3 channels are insensitive to external TEA
(5 mM) and are inhibited by 4-AP, clofilium, ChTx and E4031 (Dalby-Brown et al.
2006). At present, little pharmacological data is available for Kv7.4 channels. This
channel shows intermediate sensitivity to TEA. Retigabine or ezogabine, a Kv7.4
activator, was approved by FDA in 2011 for adjuvant therapy of partial-onset
seizures in adults (Maljevic and Lerche 2013; Gribkoff 2003; Humphries and Dart
2015; Jenkinson 2006). The other compounds inhibiting this channel include
bepridil and XE991 (Dalby-Brown et al. 2006; Augustynek et al. 2016). Kv7.5
channels are activated by BMS204352 and retigabine, and further promote conduc-
tion of the channels. TEA exerts little sensitivity towards Kv7.5 channels. XE991
also demonstrated selective inhibitory effect on Kv7.5 channels. Linopirdine is
known to block all the members of the Kv7 family (Camerino et al. 2007;
Kuzmenkov et al. 2015).
19.6 Conclusion
References
Abraham MR, Jahangir A, Alekseev AE, Terzic A (1999) Channelopathies of inwardly rectifying
potassium channels. FASEB J 13:1901–1910
Ackerman MJ, Clapham DE (1997) Ion channels—basic science and clinical disease. N Engl J Med
336:1575–1586
Adelman JP, Maylie J, Sah P (2012) Small-conductance Ca2+-activated K+ channels: form and
function. Annu Rev Physiol 74:245–269
Akopova OV (2018) Direct and off-target effects of ATP-sensitive potassium channels opener
diazoxide. J Drug Metab Toxicol 8:227
Alagem N, Dvir M, Reuveny E (2001) Mechanism of Ba2+ block of a mouse inwardly rectifying K
+ channel: differential contribution by two discrete residues. J Physiol 534:381–393
Alexander SP, Benson HE, Faccenda E, Pawson AJ, Sharman JL, Catterall WA, Spedding M,
Peters JA, Harmar AJ (2013) The concise guide to pharmacology 2013/14: ion channels. Br J
Pharmacol 170:1607–1651
Alexander SP, Striessnig J, Kelly E, Marrion NV, Peters JA, Faccenda E, Harding SD, Pawson AJ,
Sharman JL, Southan C (2017) The concise guide to pharmacology 2017/18: Voltage-gated ion
channels. Br J Pharmacol 174:S160–S194
An WF, Bowlby MR, Betty M, Cao J, Ling H-P, Mendoza G, Hinson JW, Mattsson KI, Strassle
BW, Trimmer JS (2000) Modulation of A-type potassium channels by a family of calcium
sensors. Nature 403:553
Atkinson NS, Robertson GA, Ganetzky B (1991) A component of calcium-activated potassium
channels encoded by the Drosophila slo locus. Science 253:551–555
Augustynek B, Kunz WS, Szewczyk A (2016) Guide to the pharmacology of mitochondrial
potassium channels. Pharmacol Mitochondria 240:103–127. Springer
Bearden D, Strong A, Ehnot J, Digiovine M, Dlugos D, Goldberg EM (2014) Targeted treatment of
migrating partial seizures of infancy with quinidine. Ann Neurol 76:457–461
Berg AP, Sen N, Bayliss DA (2007) TrpC3/C7 and Slo2. 1 are molecular targets for metabotropic
glutamate receptor signaling in rat striatal cholinergic interneurons. J Neurosci 27:8845–8856
Bergeron Z, Bingham J-P (2012) Scorpion toxins specific for potassium (K+) channels: a historical
overview of peptide bioengineering. Toxins 4:1082–1119
Bhattacharjee A, Joiner WJ, Wu M, Yang Y, Sigworth FJ, Kaczmarek LK (2003) Slick (Slo2. 1), a
rapidly-gating sodium-activated potassium channel inhibited by ATP. J Neurosci
23:11681–11691
Bhave G, Lonergan D, Chauder BA, Denton JS (2010) Small-molecule modulators of inward
rectifier K+ channels: recent advances and future possibilities. Future Med Chem 2:757–774
Bhave G, Chauder BA, Liu W, Dawson ES, Kadakia R, Nguyen TT, Lewis LM, Meiler J, Weaver
CD, Satlin LM (2011) Development of a selective small-molecule inhibitor of Kir1. 1, the renal
outer medullary potassium channel. Mol Pharmacol 79:42–50
Calderone V (2002) Large-conductance, Ca2+-activated K+ channels: function, pharmacology and
drugs. Curr Med Chem 9:1385–1395
Camerino DC, Tricarico D, Desaphy J-F (2007) Ion channel pharmacology. Neurotherapeutics
4:184–198
Campos Rosa J, Galanakis D, Piergentili A, Bhandari K, Ganellin CR, Dunn PM, Jenkinson DH
(2000) Synthesis, molecular modeling, and pharmacological testing of bis-quinolinium
19 Pharmacology of Potassium Channels 673
Ishihara K, Hiraoka M, Ochi R (1996) The tetravalent organic cation spermine causes the gating of
the IRK1 channel expressed in murine fibroblast cells. J Physiol 491:367–381
Jackson WF (2017) Potassium channels in regulation of vascular smooth muscle contraction and
growth. Adv Pharmacol 78:89–144. Elsevier
Jenkinson DH (2006) Potassium channels–multiplicity and challenges. Br J Pharmacol 147:S63–
S71
Jensen BS, Ødum N, JØrgensen NK, Christophersen P, Olesen S-P (1999) Inhibition of T cell
proliferation by selective block of Ca2+-activated K+ channels. Proc Natl Acad Sci
96:10917–10921
Jensen BS, Olsen S-P, Jørgensen TD, Strøbæk D, Christophersen P, Ødum N (2003) Chemical
compounds having ion channel blocking activity for the treatment of immune dysfunction.
Google Patents
Jensen MØ, Jogini V, Borhani DW, Leffler AE, Dror RO, Shaw DE (2012) Mechanism of voltage
gating in potassium channels. Science 336:229–233
Ji X-C, Zhao W-H, Cao D-X, Shi Q-Q, Wang X-L (2011) Novel neuroprotectant chiral 3-n-
butylphthalide inhibits tandem-pore-domain potassium channel TREK-1. Acta Pharmacol Sin
32:182
Jiménez-Vargas JM, Possani LD, Luna-Ramirez K (2017) Arthropod toxins acting on neuronal
potassium channels. Neuropharmacology 127:139–160
Kaczmarek LK (2013) Slack, slick, and sodium-activated potassium channels. ISRN Neurosci
2013:354262
Kaczmarek LK, Aldrich RW, Chandy KG, Grissmer S, Wei AD, Wulff H (2017) International
Union of Basic and Clinical Pharmacology. C Nomenclature and properties of calcium-activated
and sodium-activated potassium channels. Pharmacol Rev 69:1–11
Kaczorowski GJ, Garcia ML (1999) Pharmacology of voltage-gated and calcium-activated potas-
sium channels. Curr Opin Chem Biol 3:448–458
Kaczorowski GJ, Knaus H-G, Leonard RJ, McManus OB, Garcia ML (1996) High-conductance
calcium-activated potassium channels; structure, pharmacology, and function. J Bioenerg
Biomembr 28:255–267
Kasap M, Dwyer DS (2018) Two-pore domain potassium channels (K2Ps) as drug targets in
neuroinflammation. Neuroinflammation:413–427. https://doi.org/10.1016/B978-0-12-811709-
5.00022-3. Elsevier
Kaufmann K, Romaine I, Days E, Pascual C, Malik A, Yang L, Zou B, Du Y, Sliwoski G, Morrison
RD (2013) Ml297 (VU0456810), the first potent and selective activator of the GIRK potassium
channel, displays antiepileptic properties in mice. ACS Chem Nerosci 4:1278–1286
Kennard LE, Chumbley JR, Ranatunga KM, Armstrong SJ, Veale EL, Mathie A (2005) Inhibition
of the human two-pore domain potassium channel, TREK-1, by fluoxetine and its metabolite
norfluoxetine. Br J Pharmacol 144:821–829
Ketchum KA, Joiner WJ, Sellers AJ, Kaczmarek LK, Goldstein SA (1995) A new family of
outwardly rectifying potassium channel proteins with two pore domains in tandem. Nature
376:690
Kim D (2005) Physiology and pharmacology of two-pore domain potassium channels. Curr Pharm
Des 11:2717–2736
Kim GE, Kaczmarek LK (2014) Emerging role of the KCNT1 Slack channel in intellectual
disability. Front Cell Neurosci 8:209
Kim DM, Nimigean CM (2016) Voltage-gated potassium channels: a structural examination of
selectivity and gating. Cold Spring Harb Perspect Biol 8:a029231
Kim Y, Bang H, Kim D (2000) TASK-3, a new member of the tandem pore K+ channel family. J
Biol Chem 275:9340–9347
Kobayashi T, Washiyama K, Ikeda K (2004) Inhibition of G protein-activated inwardly rectifying K
+ channels by various antidepressant drugs. Neuropsychopharmacology 29:1841
Köhling R, Wolfart J (2016) Potassium channels in epilepsy. Cold Spring Harb Perspect Med 6:
a022871
19 Pharmacology of Potassium Channels 677
Kuang Q, Purhonen P, Hebert H (2015) Structure of potassium channels. Cell Mol Life Sci
72:3677–3693
Kurata HT, Phillips LR, Rose T, Loussouarn G, Herlitze S, Fritzenschaft H, Enkvetchakul D,
Nichols CG, Baukrowitz T (2004) Molecular basis of inward rectification: polyamine interac-
tion sites located by combined channel and ligand mutagenesis. J Gen Physiol 124:541–554
Kuzmenkov A, Grishin E, Vassilevski A (2015) Diversity of potassium channel ligands: focus on
scorpion toxins. Biochemistry (Mosc) 80:1764–1799
Lam J, Coleman N, Garing ALA, Wulff H (2013) The therapeutic potential of small-conductance
KCa2 channels in neurodegenerative and psychiatric diseases. Expert Opin Ther Targets
17:1203–1220
Langguth B, Elgoyhen AB, Schlee W (2016) Potassium channels as promising new targets for
pharmacologic treatment of tinnitus: can Internet-based ‘crowd sensing’initiated by patients
speed up the transition from bench to bedside? Expert Opin Ther Targets 20:251–254. Taylor &
Francis
Lawson K (2000) Potassium channel openers as potential therapeutic weapons in ion channel
disease. Kidney Int 57:838–845
Ledoux J, Werner ME, Brayden JE, Nelson MT (2006) Calcium-activated potassium channels and
the regulation of vascular tone. Physiology 21:69–78
Lehmann-Horn F, Lerche H, Jurkat-Rott K (2003) Skeletal muscle channelopathies: myotonias,
periodic paralyses and malignant hyperthermia. In: Handbook of clinical neurophysiology, vol
2. Elsevier, Amsterdam, pp 457–483
Lengyel M, Dobolyi A, CzirjáK G, Enyedi P (2017) Selective and state-dependent activation of
TRESK (K2P18. 1) background potassium channel by cloxyquin. Br J Pharmacol
174:2102–2113
Lesage F (2003) Pharmacology of neuronal background potassium channels. Neuropharmacology
44:1–7
Lesage F, Lazdunski M (2000) Molecular and functional properties of two-pore-domain potassium
channels. Am J Physiol Renal Physiol 279:F793–F801
Li X-Y, Toyoda H (2015) Role of leak potassium channels in pain signaling. Brain Res Bull
119:73–79
Longman SD, Hamilton TC (1992) Potassium channel activator drugs: mechanism of action,
pharmacological properties, and therapeutic potential. Med Res Rev 12:73–148
Lopatin AN, Makhina EN, Nichols CG (1994) Potassium channel block by cytoplasmic polyamines
as the mechanism of intrinsic rectification. Nature 372:366
Loucif AJ, Saintot PP, Liu J, Antonio BM, Zellmer SG, Yoger K, Veale EL, Wilbrey A, Omoto K,
Cao L (2018) GI-530159, a novel, selective, mechanosensitive two-pore-domain potassium
(K2P) channel opener, reduces rat dorsal root ganglion neuron excitability. Br J Pharmacol
175:2272–2283
Luzhkov V, Åqvist J (2005) Ions and blockers in potassium channels: insights from free energy
simulations. Biochim Biophys Acta 1747:109–120
Mackinnon R (2003) Potassium channels. FEBS Lett 555:62–65
Malayev A, Nelson D, Philipson L (1995) Mechanism of clofilium block of the human Kv1.
5 delayed rectifier potassium channel. Mol Pharmacol 47:198–205
Maljevic S, Lerche H (2013) Potassium channels: a review of broadening therapeutic possibilities
for neurological diseases. J Neurol 260:2201–2211
Mathie A, Wooltorton JR, Watkins CS (1998) Voltage-activated potassium channels in mammalian
neurons and their block by novel pharmacological agents. Gen Pharmacol Vasc S 30:13–24
Meadows H, Chapman C, Duckworth D, Kelsell R, Murdock P, Nasir S, Rennie G, Randall A
(2001) The neuroprotective agent sipatrigine (BW619C89) potently inhibits the human tandem
pore-domain K+ channels TREK-1 and TRAAK. Brain Res 892:94–101
Meera P, Wallner M, Song M, Toro L (1997) Large conductance voltage-and calcium-dependent K
+ channel, a distinct member of voltage-dependent ion channels with seven N-terminal
678 S. Deka et al.
(UCL 1684), the first nanomolar, non-peptidic blocker of the apamin-sensitive Ca2+-activated
K+ channel. J Med Chem 41:2–5
Rubaiy HN (2016) The therapeutic agents that target ATP-sensitive potassium channels. Acta
Pharm 66:23–34
Rudy B, Mcbain CJ (2001) Kv3 channels: voltage-gated K+ channels designed for high-frequency
repetitive firing. Trends Neurosci 24:517–526
Salata J, Jurkiewicz N, Sanguinetti M, Siegl P, Claremon D, Remy D, Elliot J, Libby B (1996) The
novel class III antiarrhythmic agent, L-735,821 is a potent and selective blocker of IKs in guinea
pig ventricular myocytes. Circulation 94:1–529
Salata JJ, Jurkiewicz NK, Wang J, Evans BE, Orme HT, Sanguinetti MC (1998) A novel
benzodiazepine that activates cardiac slow delayed rectifier K+ currents. Mol Pharmacol
54:220–230
Sanchez M, McManus O (1996) Paxilline inhibition of the alpha-subunit of the high-conductance
calcium-activated potassium channel. Neuropharmacology 35:963–968
Sánchez-Carranza O, Torres-Rodríguez P, Darszon A, Treviño CL, López-González I (2015)
Pharmacology of hSlo3 channels and their contribution in the capacitation-associated hyperpo-
larization of human sperm. Biochem Biophys Res Commun 466:554–559
Sands S, Lewis R, Cahalan M (1989) Charybdotoxin blocks voltage-gated K+ channels in human
and murine T lymphocytes. J Gen Physiol 93:1061–1074
Schneider ER, Anderson EO, Gracheva EO, Bagriantsev SN (2014) Temperature sensitivity of
two-pore (K2P) potassium channels. Curr Top Membr 74:113–133. Elsevier
Seifert G, Henneberger C, Steinhaeuser C (2018) Diversity of astrocyte potassium channels: An
update. Brain Res Bull 136:26–36
Shah NH, Aizenman E (2014) Voltage-gated potassium channels at the crossroads of neuronal
function, ischemic tolerance, and neurodegeneration. Transl Stroke Res 5:38–58
Shieh C-C, Coghlan M, Sullivan JP, Gopalakrishnan M (2000) Potassium channels: molecular
defects, diseases, and therapeutic opportunities. Pharmacol Rev 52:557–594
Sokolova O, Kolmakova-Partensky L, Grigorieff N (2001) Three-dimensional structure of a
voltage-gated potassium channel at 2.5 nm resolution. Structure 9:215–220
Staudacher I, Illg C, Chai S, Deschenes I, Seehausen S, Gramlich D, Müller ME, Wieder T, Rahm
A-K, Mayer C (2018a) Cardiovascular pharmacology of K 2P 17.1 (TASK-4, TALK-2) two-
pore-domain K+ channels. Naunyn Schmiedebergs Arch Pharmacol 391:1119–1131
Staudacher I, Seehausen S, Illg C, Lugenbiel P, Schweizer PA, Katus HA, Thomas D (2018b)
Cardiac K2P13. 1 (THIK-1) two-pore-domain K+ channels: pharmacological regulation and
remodeling in atrial fibrillation. Prog Biophys Mol Biol 144:128–138
Strøbæk D, Teuber L, JØrgensen TD, Ahring PK, Kjær K, Hansen RS, Olesen SP,
Christophersen P, Skaaning-Jensen B (2004) Activation of human IK and SK Ca2+-activated
K+ channels by NS309 (6, 7-dichloro-1H-indole-2, 3-dione 3-oxime). Biochim Biophys Acta
1665:1–5
Sturgess N, Cook D, Ashford MJ, Hales CN (1985) The sulphonylurea receptor may be an
ATP-sensitive potassium channel. Lancet 326:474–475
Swanson R, Marshall J, Smith JS, Williams JB, Boyle MB, Folander K, Luneau CJ, Antanavage J,
Oliva C, Buhrow SA (1990) Cloning and expression of cDNA and genomic clones encoding
three delayed rectifier potassium channels in rat brain. Neuron 4:929–939
Swartz KJ, Mackinnon R (1997a) Hanatoxin modifies the gating of a voltage-dependent K+
channel through multiple binding sites. Neuron 18:665–673
Swartz KJ, Mackinnon R (1997b) Mapping the receptor site for hanatoxin, a gating modifier of
voltage-dependent K+ channels. Neuron 18:675–682
Syme CA, Gerlach AC, Singh AK, Devor DC (2000) Pharmacological activation of cloned
intermediate-and small-conductance Ca2+-activated K+ channels. Am J Physiol Cell Physiol
278:C570–C581
680 S. Deka et al.
Xia X-M, Fakler B, Rivard A, Wayman G, Johnson-Pais T, Keen J, Ishii T, Hirschberg B, Bond C,
Lutsenko S (1998) Mechanism of calcium gating in small-conductance calcium-activated
potassium channels. Nature 395:503
Yamashita T, Horio Y, Yamada M, Takahashi N, Kondo C, Kurachi Y (1996) Competition between
Mg2+ and spermine for a cloned IRK2 channel expressed in a human cell line. J Physiol
493:143–156
Yang B, Gribkoff VK, Pan J, Damagnez V, Dworetzky SI, Boissard CG, Bhattacharjee A, Yan Y,
Sigworth FJ, Kaczmarek LK (2006) Pharmacological activation and inhibition of Slack (Slo2.
2) channels. Neuropharmacology 51:896–906
Yellen G (2002) The voltage-gated potassium channels and their relatives. Nature 419:35
Yeung SYM, Thompson D, Wang Z, Fedida D, Robertson B (2005) Modulation of Kv3 subfamily
potassium currents by the sea anemone toxin BDS: significance for CNS and biophysical
studies. J Neurosci 25:8735–8745
Yost CS (2000) Tandem pore domain K channels an important site of volatile anesthetic action.
Curr Drug Targets 1:207–217
Yu H-B, Li M, Wang W-P, Wang X-L (2016) High throughput screening technologies for ion
channels. Acta Pharmacol Sin 37:34
Zeng X-H, Yang C, Xia X-M, Liu M, Lingle CJ (2015) SLO3 auxiliary subunit LRRC52 controls
gating of sperm KSPER currents and is critical for normal fertility. Proc Natl Acad Sci
112:2599–2604
Zhang X, Anderson JW, Fedida D (1997) Characterization of nifedipine block of the human heart
delayed rectifier, hKv1. 5. J Pharmacol Exp Ther 281:1247–1256
Zhou B-Y, Ma W, Huang X-Y (1998) Specific antibodies to the external vestibule of voltage-gated
potassium channels block current. J Gen Physiol 111:555–563
Pharmacology of Calcium Channel
20
Santanu Mallik and Pratap Chandra Acharya
Abstract
S. Mallik
Department of Pharmacy, Tripura University (A Central University), Suryamaninagar, Tripura (W),
India
Bharat Pharmaceutical Technology, Amtali, Agartala, Tripura (W), India
P. C. Acharya (*)
Department of Pharmacy, Tripura University (A Central University), Suryamaninagar, Tripura (W),
India
e-mail: [email protected]
Keywords
Abbreviations
20.1 Introduction
Erwin Neher and Bert Sakmann, Nobel laureates (1970), invented the technique
“patch clamp” (De la Peña and Gomis 2019) which confirmed the practical existence
of ion channels in a physiological environment. Ion channels are transmembrane
glycoprotein pores having the open and close properties like gates. These channels
allow the movement of ions in a regulated manner. Electrochemical gradient governs
the direction and rate of ion movement. The transportation is always down the
gradient and the speed of ion transportation through the channels is very high
which is approximately about 107 ions/s (Sanchez-Sandoval and Gomora 2019;
Uzieliene et al. 2018). The types of calcium channels are shown in Tables 20.1
and 20.2 (Zamponi et al. 2015; Spedding and Paoletti 1992).
In the recent medical science, diseases like migraine, epilepsy, hypertension, and
cerebral ischemia and rarer diseases like night blindness have been treated by
targeting calcium channels. The channels allow calcium ions to flow out of the
endoplasmic reticulum or to flow into the cell (Zamponi et al. 2010; Marchetti 2013).
The opening of such ion channels is initiated due to the changes in voltage across the
membrane, for example, signal propagation of nerve cells. One more factor which
helps open the channel is the ligand molecule like inositol 1,4,5-triphosphate (IP3)
(Prakriya and Lewis 2015; Simms and Zamponi 2014). The pancreatic beta cells,
neurons, myocytes, etc., have calcium channels. Such kind of channels forms most of
the systematic molecular connection between biochemical signaling intracellularly
20 Pharmacology of Calcium Channel 685
Fig. 20.1 Pictorial representation of α1, β, α-2δ and γ subunits of calcium channel. Four homolo-
gous domains of α1 subunit (1–4) with helical structures have been presented
The opening and closing of the calcium channels are the fundamental properties.
Depending on the mechanism of these properties, calcium channels are categorized
(Fig. 20.1) under two broad headings: (1) voltage dependent and (2) receptor
operated. Voltage-dependent calcium channels are further subclassified depending
on the pharmacological sensitivities, kinetic properties, and voltage sensitivities
(Zamponi 2016).
It has been reported that L- and T-type channels are present in skeletal and cardiac
muscles where N-type channel is absent. In sympathetic neurons of ganglia, T-type
is not present but both L- and N-types are available. The T-, L-, and N-types of
receptors coexist in dorsal root ganglia sensory neurons. Furthermore, differences
have been observed in the same type of channel located at different organs. For
instance, L-type calcium channel present in the skeletal muscle and heart shows
differences in electrophysiological and pharmacological properties (Kappel et al.
2013; Rose et al. 2013; Li et al. 2017a, b).
Receptor-operated calcium channels or calcium channels operated by receptor-
dependent mechanism are usually opened after receiving signals from an activated
associate receptor (Fig. 20.2). Chloride and ion channels are two major types
20 Pharmacology of Calcium Channel 687
Drug affinity entirely depends on the specific gating status of the channels for the
drugs that target ion channels. State dependency is the reason behind the clinical
utility of drug molecules. On the other hand, experimental use of a drug is an
evidential proof of successful state-dependent inhibition. It is very important to
contemplate hypothetical considerations of state-dependent inhibition, which
includes drug-inactivation synergism, use-dependent block (the rate of nerve stimu-
lation is proportional to the degree of block), guarded receptors (acyclic), and cyclic
or allosteric receptor (Yoder et al. 2018).
The idea of drug binding was introduced by Armstrong in 1966 and Strichartz in
1973. Different strengths to resting closed (R), open (O), and closed inactivated
(I) ion channels have been reported. Later on, the idea was accepted as the theoretical
concept of a modulated drug receptor (Zaydman et al. 2014; Lenaeus et al. 2017).
688 S. Mallik and P. C. Acharya
Depending on this hypothesis, study on the biological phenomena with the help of
physics and drug or medication action experiments on ion channels helped the
medical science understand in a better way about the repression of signals of local
anesthetics and sodium channel inhibitors such as the antiarrhythmic drugs,
antiepileptics, and actions of other drugs (Borowicz and Banach 2014; Tikhonov
and Zhorov 2017).
The three conformational states of calcium channels have been widely accepted: R
state or resting, nonconducting; O state or open, conducting, or excited; and I state or
inactivated, nonconducting. During the depolarization process, channels move to I
through O from R and again back to the R state during the change in cell’s membrane
potential (hyperpolarization) (Alexander et al. 2015/16).
Fig. 20.3 Hypothetical open channel block model. (a) Open state model depicting kinetic scheme
of calcium channel, (b) molecular mechanism involved in an open state, (c) changes in kinetics of
use-dependent and open channel inhibition in reference to control, and (d) model graphical
representation of normalized current versus membrane potential in open state kinetics
Fig. 20.4 Hypothetical model of inactivated channel block. (a) Inactivated state model depicting
selective state transition kinetic scheme of calcium channel, (b) selective inhibition mechanism
involved in an inactivated state, (c) current kinetics of use-dependent and inactivated channel
inhibition in reference to control where D1 represents the initial impact of current on drugs in
comparison to control, and (d) model graphical representation of normalized current versus
membrane potential in inactivated state channel block kinetics
For decades, treatment of hypertension and myocardial ischemia has been based on
the L-type calcium channel blockers. They are the first-line choice for the above
health issues and first choice for the physicians. The arterial vasodilators
20 Pharmacology of Calcium Channel 691
resting, and inactivated conditions which is also known as modulated receptor model
usually interferes with bound drugs (Bain 2019).
Dihydropyridines, uncharged, stabilize primarily and prevail upon inactivated
channel states. Inactivated channel conformation shows higher affinity and thus
decreases with increased availability of inactivated channel states at voltage-
dependent block or more depolarized membrane potentials (Zhao et al. 2019)
which is favored by the open channel state and always favors the access of
benzothiazepines and phenylalkylamines. Blocking of pore directly together with
slowed recovery from inactivation with stabilization of inactivated channel state
results in pronounced frequency or use-dependent inhibition.
Dihydropyridines which are Ca2+ channel activators also exist. For different
reasons, the sensitivity of L-type calcium channels for dihydropyridine Ca2+ channel
blockers varies in different tissues (Surmeier et al. 2019). Variable contribution of
these L-type calcium channels to total L-type current is one of the prominent
explanations. Cav1.4 and Cav1.3 exhibit about 5- to 10-fold down to
dihydropyridines than Cav1.2, which is demonstrated as negative membrane
potentials in heterologous expression systems (Ortner et al. 2014; Kang et al.
2012). Sinoatrial node is usually dominated by Cav1.3 which can explain the
relatively weak inhibition of L-type pacemaker currents (Mesirca et al. 2015).
L-type current in substitute splicing of α1 subunits is another important factor
affecting dihydropyridine sensitivity. It has been expositional that dihydropyridines,
at lower concentrations, inhibit currents in arterial smooth muscle than in the
functional myocardium for Cav1.2. The presence of dihydropyridine-sensitive splice
variants predominantly expressed in arterial smooth muscle has been established by
rigorous analysis of Cav1.2 α1 splice variants present in heart and smooth muscles.
Moderate negative voltage activates some of these splice variants and brings out a
steady-state Ca2+ window or inward current nearer to the resting potential of cardiac
muscles (Li et al. 2017a, b; Fan et al. 2005). More amount of depolarized resting
membrane potential available in smooth muscle usually favors inactivated channels
obstructed by dihydropyridines in comparison to cardiomyocytes (Liao and Soong
2010).
Another way to enhance dihydropyridine sensitivity is that few of such splice
variants are liable to more promised inactivation of steady state. There is also
evidence that the drug binding domain 5s is affected by alternative splicing of
Cav1.2 α1 and therefore the dihydropyridines can access the inactivated channels.
Dihydropyridine sensitivity of Cav1.3 is slightly affected by alternative splicing in
the C-terminus (Jang et al. 2013).
seems to be clinically fruitful in patients with heart failure, in which elevated heart
rate has been noticed may be due to increased sympathetic drive along with
aldosterone which may be caused by secondary aldosteronism (Gordan et al.
2015). Heart rate at high level is always a risk factor in heart failure. But lowering
of heart rate with specificity may be improved by cardiovascular outcomes with the
hyperpolarization-activated cyclic nucleotide-gated channel blocking bradycardia
agent ivabradine (Tse et al. 2017).
Physiological functions controlled by different L-type calcium channel isoforms
identify these types of channels as new drug targets. It is a matter of high con-
cern because the nonselective channel blockers are in clinical use for a long time.
Furthermore, Cav1.3 selective Ca2+ channel blockers have high therapeutic potential
for many indications including neuropsychiatric disorders (Lu et al. 2015).
Peptide toxins that are collected from the venoms of organisms like fish-hunting
molluscs, spiders, and scorpions can inhibit voltage-gated calcium channels. For
example, polypeptide ω -agatoxin, subtype IVA, having a molecular weight of 5210
kDa, collected from the venom of the North American funnel web spider
Agelenopsis aperta, inhibits Cav2.1 channels (Lian et al. 2014). If they are carefully
regulated with compounds that normalize deviant gain of function, Cav2.1 channels
are not considered as good pharmacological targets in large extent where Cav2.3
channel inhibitors could potentially have an influential effect in pain (Duda et al.
2016) and seizure disorders (Kim et al. 2015).
These channels lack specific small organic inhibitors. The inhibitor of Cav2.3
channels, SNX-482 spider toxin, isolated from the venom of the spider
Hysterocrates gigas also targets Cav1.2 L-type calcium channels with A-type K+
currents. SNX-482 is also known as ω-theraphotoxin-Hg1a, or ω-TRTX-Hg1a
(Zamponi 2016; Moutal et al. 2017; Rousset et al. 2015).
occurs due to proline and mutagens of a single glycine residue in the 1326-amino-
acid position of the channel. An ensuing study reveals that ω-conotoxins MVIIA and
CVID cause changes in both the extent of inhibition and kinetics of the
co-expression of Cav2.2 δ subunit channels (Wang et al. 2016; Mollica et al. 2015).
Various peptide toxins are highly selective and are high-affinity blockers to
several Ca2+ channel subtypes, but they cannot cross the blood–brain barrier. A lot
of pore-blocking conotoxins do not perform effectively as state-dependent blockers,
namely, local anesthetics and anticonvulsants (Norton 2017; Bourinet and Zamponi
2017; Duggan and Tuck 2015). But the above issue can be overcome by increasing
affinity and selectivity of developing small organic blocking agents. The
peptidylamines are designed to mimic the pore-blocking activity of the larger ω-
conotoxin molecules. This is executed by linking acid group of N,N-disubstituted
leucine to amine group of tyrosine. The high-affinity block of Cav2.2 channels has
been evidenced in various literatures. Moreover, phenylalanine and
benzyloxyaniline derivatives have strong affinity toward Cav2.2 channel blockers,
and management of pain has been reported with scientific evidences (Jin et al. 2013;
Robinson et al. 2017; Brady et al. 2013).
Antipsychotics with D2 dopamine receptor-blocking activity are examples of few
important Cav2.2 channel blockers (Siafis et al. 2018) (Fig. 20.6, pathway 2), which
contain a core morpholine, piperidine, and/or piperazine structure and usually linked
to 1 or 2 diphenyl moieties through alkyl chains. Such agents are renowned for their
blocking property of N-type channels (Brimblecombe et al. 2015). Furthermore,
several major compounds falling under this section have been confirmed for the
management of pain in animal models. The dependency and intoxicating narcotic
properties of ethanol are also abated, and the derivatives like pyrazole piperidines
and amino-piperidine sulfonamide showed mixed action on Cav2.2 and Cav2.3
channels (François et al. 2014; Striessnig et al. 2015; Striessnig et al. 2014).
Cilnidipine is a drug which comes under dihydropyridines and acts as a blocker of
L-type calcium channel. At the same time, it is also reported that some other drugs of
the same class can also block N-type calcium channels with high accord. These
drugs have shown analgesic properties in rats and kidney protective as well as
antihypertensive properties in human volunteers. Such favorable effects of drugs
may be imputable to their action on the sympathetic nervous system of N-type
channels (Shetty et al. 2013; Adake et al. 2015; Manthri et al. 2015).
Moreover, other examples of Cav2.2 channel blockers found in the literature
include (3R)-5-(3-chloro-4-fluorophenyl)-3-methyl-3-(pyrimidin-5-ylmethyl)-1-
(1H-1,2,4-triazol-3-yl)-1,3-dihydro-2H-indol-2-one (TROX-1) which is an oxindole
compound. This compound has analgesic properties and is a state-dependent inhibi-
tor. Further, long-chain aliphatic monoamines (Beedle and Zamponi 2000) and
farnesol also block (Roullet et al. 1999) Cav2.2 channels with stronger accord and
reveal favored blockade of inactivated channels (Nimmrich and Eckert 2013; Page
et al. 2016; Ripsch et al. 2012).
The inhibitions of Cav2.2 channels are also done by other classes of
pharmacophores indicating that these channels have promising pharmacotherapeutic
implications. Left side shift has been observed in the steady-state inactivation curve
20 Pharmacology of Calcium Channel 697
Fig. 20.5 Pathway 1 represents the modulation of drugs, toxins, and signals. L-type calcium
channel active drugs are shown here. The potency of L-type channel blockers depends on mem-
brane potential as shown in pathway 2. Different signaling pathways like G-protein (pathway 2a) or
enzymes activated by G-protein-coupled receptor (GPCR) (pathway 2b) or receptor tyrosine
kinases (RTKs) (pathway 2c)
been designed for better therapeutic response (Cai et al. 2018; Ren et al. 2019; Liu
et al. 2018; Bowery 2016).
Overall, Cav2.2 channels are very important effectors of 7-transmembrane helix
receptors. The physiological importance of this regulation can be clearly exemplified
in the primary afferent pain pathway. The GPCR agonists can also regulate their
downstream effects in many other physiological processes via Ca2+ channels.
This process is connected to the entry of calcium that plays crucial role in cellular
20 Pharmacology of Calcium Channel 699
As the peptide toxins are unsuccessful in crossing the blood–brain barrier, they are
not administered orally as therapeutic agents. Kurtoxin has high affinity to inhibit
Cav3.1 calcium channels. This peptide is extracted from the venom of the scorpion
species Parabuthus transvaalicus. The above compound functions as a gating
modifier in a manner comparable to that described for the ω -agatoxin IVA, the
P-type channel blocker. Again, kurtoxin targets both N- and L-type calcium channel
700 S. Mallik and P. C. Acharya
Fig. 20.6 N-type channels regulated by toxins, drugs, and various signals. Pathway 2 indicates the
most pertinent group of active drugs. A different type of signaling pathway has been shown in
pathway 3 representing the CRMP-2 (scaffolding of proteins)
20 Pharmacology of Calcium Channel 701
isoforms and also shows its effects on sodium channel (Bourinet and Zamponi 2017;
King et al. 2008; McDonough 2007; Mary et al. 2018).
The structure of kurtoxin shows similarities to other scorpion toxins. However,
the unique surface properties of kurtoxin are the main reason for its action on T-type
channels. Two additional Parabuthus transvaalicus scorpion toxins (KLI and KLII)
also exert blocking effects on T-type calcium channels. Both toxins have weak
impact on Cav3.3 channels but can easily block sodium channels and T-type
channels (Antal and Martin-Caraballo 2019; Nanclares et al. 2018; Housley et al.
2017).
The most prominent sodium channel inhibitors are protoxins I and II which are
peptides collected from the Thrixopelma pruriens tarantula. Subtype-dependent
Cav3 channels are blocked by both peptides. Protoxin I advantageously blocks
Cav3.1 channels over Cav3.3 and Cav3.2 channels. Having the best accord for
Cav3.2 channels, protoxin II favors to act as a gating modifier. PsPTx3 is another
peptide spider toxin, extracted from Theraphosidae tarantula, which has properties to
block T-type calcium channels and perceptible selectivity for Cav3.2 channels (Kyle
et al. 2017; Bladen et al. 2014a, b; Klint et al. 2012).
In overall comparison with N-type calcium channels, peptide toxin impedes Cav3
channels which remain relatively limited and derived mostly from arachnids. Most
importantly, peptide blockers of T-type calcium channels should not be restricted to
those derived from venomous species. For instance, an intrinsic agonist of the
chemokine receptor CCR-2 (C-C motif chemokine receptor 2), monocyte
chemoattractant protein-1, can directly and actively inhibit Cav3.2 T-type calcium
channels (Ellinor et al. 1994; Silva et al. 2018; Bellampalli et al. 2019).
There are no shortfalls of T-type organic calcium channel blockers when compared
with peptide toxins. Various categories of T-type calcium channel blockers were
brought to light in different time frame (Fig. 20.7). The drug amiloride is one of the
very first accepted T-type calcium channel blockers which acts as diuretics, blocking
the Cav3.2 channels of about a single degree of magnitude with higher accord
compared to Cav3.3 and Cav3.1 channels. But it is a fact that amiloride is not a
distinct T-type calcium channel inhibitor. The antiepileptic agent ethosuximide
comes under the succinimide group. This compound displays state-dependent inhi-
bition and is a low-affinity blocker of all three categories of Cav3 channel isoforms
(Zamponi and Diaz 2017; Striessnig et al. 2015; Seo et al. 2007).
Due to the pretended selective inhibition of mibefradil, a T-type calcium channel,
initially it generated a significant excitement in this field. The United States Food
and Drug Administration (US-FDA) had approved the drug earlier for the treatment
of hypertension. But later on, it was reported across the world that the drug is
metabolized by cytochrome P450 and had severe drug–drug interactions. Hence,
mibefradil was withdrawn from the market. Few years later, a derivative of
mibefradil (NNC-55-0396) was developed which has very less interaction with
cytochrome P450 3A4 (CYP3A4) (Oshima et al. 2005; Wang et al. 2017; Okuyama
et al. 2018).
T-type calcium channels react with synthetic cannabinoid and endocannabinoid
receptor ligands. Anandamide, a neurotransmitter, and its derivative NAGly
(N-arachidonylglycine) arbitrate mighty inhibition of Cav3 calcium channels. A
synthetic carbazole derivative, NMP-7, acts as an agonist of cannabinoid receptors.
The aforesaid compound and its derivatives usually involve in blocking of T-type Ca
channels (Nam 2018; Qian et al. 2017).
Neuroleptic drugs like diphenylbutylpiperidines are familiar for their actions as
D2 dopamine receptor antagonists. Numerous members of this class of compounds
like penfluridol and pimozide significantly inhibit Cav3 channels. Major drug
20 Pharmacology of Calcium Channel 703
Fig. 20.7 Pictorial representation of different modulatory inhibitions of T-type calcium channels
like kurtoxin. The figure shows that inhibitors may block the passage physically or by binding the
gating mechanism (pathway 1). On the other hand, pathway 2a represents the direct regulation of
calcium channel by activation of GPCRs or indirect regulation by CaMKII protein kinase (pathway
2b). The channel degradation may occur due to interference of isopeptidase T or also known
as USP5 (ubiquitin-specific peptidase 5) (pathway 3) (Zamponi et al. 2015; Watanabe et al. 2015)
704 S. Mallik and P. C. Acharya
protein kinases, and phosphatases. Thus, the “kiss of death” process for a protein,
that is, ubiquitinating and deubiquitinating enzyme (DUB), namely, isopeptidases,
deubiquitinases, and ubiquitin proteases, controls Cav3.2 channels, and the T-type
channel regulation can possibly be used as a therapeutic approach for the manage-
ment of pain (François et al. 2015; Joksimovic et al. 2018).
The α2δ subunit’s molecular mechanism of action studies indicate that pregabalin
and gabapentin produce very little or zero acute retardation of calcium currents in
neuronal cell bodies transfected cells (Offord and Isom 2016). But there are few
evidences of small acute inhibitory effects. The research shows that gabapentin is
unable to inhibit calcium currents in model mouse dorsal root ganglion neurons. The
current in dorsal root ganglions from α2δ-1 is overexpressed in mice and inhibited
by gabapentin (Tano et al. 2019).
It is also found that persistent application of the above drug markedly reduces
calcium currents generated by several different α1/β/α2δ subunit combinations.
During the use of α2δ-1 or α2δ-2, the effect has been observed (Kazim 2017).
However, in the mutant α2δ, α2δ, and α2δ-3 subunits the effect does not occur
and also equally true for the subunits which bind very poorly with gabapentin. This
type of evidences strongly indicates that effects of gabapentin are in fact occurring
through binding to α2δ subunits (Freynhagen et al. 2016; Ikeda et al. 2018; Heyes
et al. 2015).
The alternative splice variant of α2δ-1 (ΔA+BΔC) shows a reduced accord for
gabapentin, whose ion is increased after nerve injury in dorsal root ganglion neurons.
This indicates the possibility of variation in the expression extent of splice variants in
neuropathic pain of individual patients (Patel and Dickenson 2016; Brockhaus et al.
2018).
The binding site for the antiepileptic drugs like gabapentin and pregabalin is
considered to be at α2δ-1 and α2δ-2 subunits. These compounds are intended to
increase the activation of GABA 2 receptor and synthesized to be immutable
lipophilic analogs. Factually gabapentin does not affect GABA levels or GABA
receptors. However, gabapentin is effective in few cases of epilepsy that are
characterized by loss of consciousness and convulsions in human and other
706 S. Mallik and P. C. Acharya
The impact of α2δ ligand drugs on transmission of synapse and release of transmitter
is a very important subject of discussion in the context of pharmacology. The
presynaptic terminals are strongly expressed by α2δ-1 subunits. However, pain
pathways can be explained by the observation of chronic effects of gabapentinoid
drugs, and the drastic effects of gabapentinoid drugs to impede release of transmitter
and synaptic transmission have been noticed in few in vitro systems (Uchitel et al.
2010; Patel et al. 2000). Deficiency of gabapentinoid drugs has acute effect on
somatic calcium currents in various systems. But such drugs are believed to have
differential effects on presynaptic terminal calcium currents in comparison to cell
bodies. As a result, more rapid inhibition of ion occurs at that stage.
Therefore, calcium channel trafficking has been noticed prominently due to the
higher turnover of calcium channel in presynaptic terminals than in somata.
Depending on the reciprocity between these different processes and presynaptic
sensitization, gabapentinoid drugs might act rapidly or more slowly. Activation of
protein kinase C is an accurate example of this phenomenon (Rogawski and Bazil
2008; Gong et al. 2018).
The very first source of Caβ subunits was identified in the purified skeletal muscle
voltage-gated calcium channel complex, and the gene for Cavβ1 was cloned later
on. As the time proceeded, Cavβ2, Caβ3, and Caβ4, three more calcium subunit
genes, were identified by similar studies. Cytoplasmic proteins or the Cavβ subunits
participate in the binding process with high accord to the intracellular loop in
between domains I and II of the Cav1 and Cav2 α1 subunits. The above binding
motif is termed as the α-interaction territory which is actually an 18-amino-acid
region in the proximal part of the I–II linker (Miriyala et al. 2008; Jangsangthong
et al. 2011; Findeisen et al. 2017).
Cavβ subunits having a protected src homology-3 domain (or SH3 domain) were
found by homology modeling processes. This subunit is also found in guanylate
kinase like domain which is interconnected by a flexible loop. Due to the mutations
708 S. Mallik and P. C. Acharya
in the active site, the kinase activity is zero in the domain of guanylate kinase. There
are three studies which have resolved the crystal structure of the conserved domains
in various β subunits (Newman and Prehoda 2009; Stölting et al. 2015). The study
showed that the AID (α-interaction domain) peptide interaction site is in a groove in
the domain like guanylate kinase. The α-helical structure of the α-interaction domain
in the intact I–II loop is inflicted by binding to the Caβ subunit. This is predicted to
continue till the end of S6 in transmembrane domain I. In this way, β subunits are
considered as safeguards to induce folding of the α1 subunit with accuracy
(Hofmann et al. 2015; Kazim 2017; Hidalgo et al. 2019).
Most common and rare diseases like epilepsy, cardiac dysfunction, and others are
connected to Cavβ subunit. Hypothetical consideration about designing of a drug
targeting the indentation within Cavβ into which the α-interaction domain peptide is
incorporated could impede the interaction between the β subunits and Cavα1. In this
way, it reduces the function of calcium channel, which on the other hand is beneficial
in certain medical conditions such as hypertension and chronic pain (Dolphin 2016;
Felix and Weiss 2017).
However, the interactivity between Cavβ and the alpha-interacting domain region
is of very high accord and includes a number of remnants. It is difficult to extract out
the process through which small molecules compete for conjugation and the selec-
tivity of different Cavα1 and β subunits. The use of cell-penetrating peptides
describes the interference between the interaction between Cav2.2 and collapsin
response mediator protein (Angelini 2015; Butcher et al. 2006; Korkosh et al. 2019).
To study the specific roles for different calcium channel isoforms, a number of
splice variants and supplementary subunits have been identified, aided by existence
of human mutations and knockout mouse models of these channels and their
supplementary subunits. Physiological activities that are controlled by Cav1.2 and
Cav1.3 are identified through different procedures. Both channels act as potentially
novel drug targets if selectivity can be achieved (Dolphin 2016; Thalhammer et al.
2017). For the treatment of hypertension, nonselective blockers of these channels
have been used and their profiles of adverse effect have been studied very closely.
Depolarized potentials have been found in vascular Cav1.2, and due to this, the
selectivity of dihydropyridines is nevertheless attained in vivo for aiming the tissue.
It is intended to bind with greater accord to inactivated channels. Few specific drugs
have promising therapeutic efficacy for selective Cav1.3 blockers with various
indications like neuroprotection, neuropsychiatric diseases, Parkinson’s disease,
and resistant hypertension associated with hyperaldosteronism (Ross 2018; Torres
et al. 2015).
Ensuring novel classes of Cav2.2 channel blockers with enormous selectivity,
accord, and use dependence is a challenge for the researchers. Ziconotide (peptide ω -
20 Pharmacology of Calcium Channel 709
conotoxin MVIIA) has been administered intrathecally for use in intractable pain.
The Cav2.2 channel blockers which act as analgesics might also be effective in
conditions such as drug anxiety and dependence. The Cav3 (T-type) channels are
important regulators of pacemaker activity and neuronal firing and play crucial roles
in the cardiovascular system (Elies et al. 2016; Woon and Balijepalli 2015). Their
dysfunction contributes to a number of chronic complications such as epilepsy and
pain. In this way, they are both potential and actual drug targets for such medical
conditions, namely, ethosuximide. But due to the protection of absence seizures by
ethosuximide, the present-day clinical use of many types of such promising T-type
channel blocking small organic molecules has been largely restricted (Bourinet et al.
2016; Lee et al. 2019).
The Cav3 auxiliary subunit α2δ1 is a well promising drug target for pregabalin
and gabapentin and has been used in chronic neuropathic pain. When combined with
other drugs in several forms, it is useful in the treatment of epilepsy. The drug also
shows binding interest with α2δ2, but not α2δ3. A selective or more potent α2δ1
ligand would have less side effects, and whether a similar ligand targeting α2δ3
might be of therapeutic use is still under investigation (Choi et al. 2016; Rui et al.
2016; Chew and Khanna 2018).
But few specific calcium channel blockers are likely to hold eminent promises
and hope for therapeutic involvement in coming future.
20.7 Conclusions
Different types of calcium ion channels like L-type, T-type, and N-type found in
muscle cells, neurons, or endocrine cells can be easily distinguished by their
pharmacological responses. All the three types of ion channels are available in
dorsal root ganglion with different activation threshold and identical selectivity.
Animal studies reveal that these ion channels can regulate the use of targeted drug
delivery effectively. The Cav1.2 and Cav1.3 subunits have strong indications of
enhancing selectivity of novel drugs, whereas nonselective dihydropyridines and
other types of calcium channel blockers have been used for the treatment of elevated
blood pressure for decades. The drug ziconotide, a peptide ω -conotoxin MVIIA, is
popularly used in unmanageable pain of individuals through intrathecal route and
acts by blocking of Cav2.2 channel.
In modern times, the management of epilepsy and chronic neuropathic pain has
been widely controlled by targeting α2δ-1 subunit with minimal side effects. Drugs
like pregabalin and gabapentin usually bind to the ligand binding site to exert
therapeutic activity.
The Cav3 or T-type channels are connected with the cardiovascular system by
regulating the action of pacemaker. Furthermore, it is essential to understand their
interaction with anandamide, arachidonic acid, redox agents, and G-proteins. The
randomized clinical investigation further indicates the promising use of Cav3 ligands
in various disease management related to pain, heart, hypertension. In a nutshell, cal-
cium channel blocking agents, depending on the selectivity and mechanism of their
710 S. Mallik and P. C. Acharya
pharmacological action, are going to hold great commitment for therapeutic arbitra-
tion in the coming days.
References
Adake P, Somashekar HS, Rafeeq PM, Umar D, Basheer B, Baroudi K (2015) Comparison of
amlodipine with cilnidipine on antihypertensive efficacy and incidence of pedal edema in mild
to moderate hypertensive individuals: a prospective study. J Adv Pharm Technol Res 6(2):81
Adams DJ, Berecki G (2013) Mechanisms of conotoxin inhibition of N-type (Cav2. 2) calcium
channels. Biochim Biophys Acta 1828(7):1619–1628
Adams DJ, Callaghan B, Berecki G (2012) Analgesic conotoxins: block and G protein-coupled
receptor modulation of N-type (CaV2. 2) calcium channels. Br J Pharmacol 166(2):486–500
Aggarwal S, Loomba RS, Arora RR (2016) Effects of concurrent calcium channel blocker on
antiplatelet efficacy of clopidogrel therapy: a systematic review. Am J Ther 23(1):e29–e36
Akanuma SI, Yamakoshi A, Sugouchi T, Kubo Y, Hartz AM, Bauer B, Hosoya KI (2018) Role of
L-type amino acid transporter 1 at the inner blood-retinal barrier in the blood-to-retina transport
of gabapentin. Mol Pharm 15(6):2327–2337
Alexander SP, Peters JA, Kelly E, Marrion N, Benson HE, Faccenda E, Pawson AJ, Sharman JL,
Southan C, Davies JA (2015) The Concise Guide to pharmacology 2015/16: ligand-gated ion
channels. Br J Pharmacol 172(24):5870–5903
Angelini M (2015) Role of CLIC1 and L-type calcium channels in the pathophysiology of
glioblastoma and ventricular arrhythmias. PhD Thesis, University of California Los Angeles,
The United States of America
Antal L, Martin-Caraballo M (2019) T-type calcium channels in cancer. Cancer 11(2):134
Arsene CG, Kaiser P, Paleari R, Henrion A, Spannagl M, Mosca A (2018) Determination of HbA2
by quantitative bottom-up proteomics and isotope dilution mass spectrometry. Clin Chim Acta
487:318–324
Ataei S, Abaspanah S, Haddadi R, Mohammadi M, Nili-Ahmadabadi A (2019) Therapeutic
potential of dihydropyridine calcium channel blockers on oxidative injury caused by
organophosphates in cortex and cerebellum: an in vivo study. Indian J Clin Biochem 1–8.
https://doi.org/10.1007/s12291-019-00830-3 (Article in press)
Badou A, Jha MK, Matza D, Flavell RA (2013) Emerging roles of L-type voltage-gated and other
calcium channels in T lymphocytes. Front Immunol 4:243
Bain A (2019) Use of calcium channel blockers in cardiovascular disease. Br J of Cardiac Nurs 14
(2):64–70
Bajaj S, Han J (2019) Venom-derived peptide modulators of cation-selective channels: friend, foe
or frenemy. Front Pharmacol 10:58
Baloh RW (2019) Promising new treatments for chronic pain. In: Sciatica and chronic pain.
Springer, Cham, pp 109–120
Barua S, Kim JY, Kim JY, Kim JH, Lee JE (2019) Therapeutic effect of agmatine on neurological
disease: focus on ion channels and receptors. Neurochem Res 44(4):735–750
Beedle AM, Zamponi GW (2000) Block of voltage-dependent calcium channels by aliphatic
monoamines. Biophys J 79(1):260–270
Bellampalli SS, Ji Y, Moutal A, Cai S, Wijeratne EK, Gandini MA, Yu J, Chefdeville A, Dorame A,
Chew LA, Madura CL (2019) Betulinic acid, derived from the desert lavender Hyptis emoryi,
attenuates paclitaxel-, HIV-, and nerve injury–associated peripheral sensory neuropathy via
block of N-and T-type calcium channels. Pain 160(1):117–135
Bidaud I, Mezghrani A, Swayne LA, Monteil A, Lory P (2006) Voltage-gated calcium channels in
genetic diseases. Biochim Biophys Acta 1763(11):1169–1174
20 Pharmacology of Calcium Channel 711
Bladen C, Gündüz MG, Şimşek R, Şafak C, Zamponi GW (2014a) Synthesis and evaluation of
1, 4-dihydropyridine derivatives with calcium channel blocking activity. Pflügers Arch 466
(7):1355–1363
Bladen C, Hamid J, Souza IA, Zamponi GW (2014b) Block of T-type calcium channels by
protoxins I and II. Mol Brain 7(1):36
Bogeski I, Kappl R, Kummerow C, Gulaboski R, Hoth M, Niemeyer BA (2011) Redox regulation
of calcium ion channels: chemical and physiological aspects. Cell Calcium 50(5):407–423
Borowicz KK, Banach M (2014) Antiarrhythmic drugs and epilepsy. Pharmacol Rep 66
(4):545–551
Bourinet E, Zamponi GW (2017) Block of voltage-gated calcium channels by peptide toxins.
Neuropharmacology 127:109–115
Bourinet E, Stotz SC, Spaetgens RL, Dayanithi G, Lemos J, Nargeot J, Zamponi GW (2001)
Interaction of SNX482 with domains III and IV inhibits activation gating of alpha
(1E) (Ca(V)2.3) calcium channels. Biophys J 81(1):79–88
Bourinet E, Francois A, Laffray S (2016) T-type calcium channels in neuropathic pain. Pain 157:
S15–S22
Bowery NG (2016) Baclofen: therapeutic use and potential of the prototypic GABA B receptor
agonist. In: GABAB receptor. Humana Press, Cham, pp 337–356
Brady R, Baell J, Norton R (2013) Strategies for the development of conotoxins as new therapeutic
leads. Mar Drugs 11(7):2293–2313
Brimblecombe KR, Gracie CJ, Platt NJ, Cragg SJ (2015) Gating of dopamine transmission by
calcium and axonal N-, Q-, T- and L-type voltage-gated calcium channels differs between
striatal domains. J Physiol 593:929–946
Brockhaus J, Schreitmüller M, Repetto D, Klatt O, Reissner C, Elmslie K, Heine M, Missler M
(2018) α-Neurexins together with α2δ-1 auxiliary subunits regulate Ca2+ influx through CaV2.
1 channels. J Neurosci 38(38):8277–8294
Bruckner JJ, Zhan H, Gratz SJ, Rao M, Ukken F, Zilberg G, O’Connor-Giles KM (2017) Fife
organizes synaptic vesicles and calcium channels for high-probability neurotransmitter release. J
Cell Biol 216(1):231–246
Buchta WC, Moutal A, Hines B, Garcia-Keller C, Smith AC, Kalivas P, Khanna R, Riegel A
(2019) Dynamic CRMP2 regulation of CaV2. 2 in the prefrontal cortex contributes to the
reinstatement of cocaine seeking. bioRxiv, 533083
Butcher AJ, Leroy J, Richards MW, Pratt WS, Dolphin AC (2006) The importance of occupancy
rather than affinity of Cavβ subunits for the calcium channel I–II linker in relation to calcium
channel function. J Physiol 574(2):387–398
Cai F, Xu N, Liu Z, Ding R, Yu S, Dong M, Wang S, Shen J, Tae HS, Adams DJ, Zhang X (2018)
Targeting of N-Type calcium channels via GABAB-receptor activation by α-Conotoxin Vc1.
1 variants displaying improved analgesic activity. J Med Chem 61(22):10198–10205
Caricati-Neto A, Bergantin LB (2016) New therapeutic strategy of Alzheimer’s and Parkinson’s
diseases: pharmacological modulation of neural Ca/cAMP intracellular signaling interaction
2. Asian J Pharm Pharmacol 2(6):136–143
Carstens BB, Clark RJ, Daly NL, Harvey PJ, Kaas Q, Craik DJ (2011) Engineering of conotoxins
for the treatment of pain. Curr Pharm Des 17(38):4242–4253
Cassidy JS, Ferron L, Kadurin I, Pratt WS, Dolphin AC (2014) Functional exofacially tagged
N-type calcium channels elucidate the interaction with auxiliary α2δ-1 subunits. Proc Natl Acad
Sci 111(24):8979–8984
Catterall WA, Swanson TM (2015) Structural basis for pharmacology of voltage-gated sodium and
calcium channels. Mol Pharmacol 88(1):141–150
Celli R, Santolini I, Guiducci M, Van Luijtelaar G, Parisi P, Striano P, Gradini R, Battaglia G,
Ngomba TR, Nicoletti F (2017) The α2δ subunit and absence epilepsy: beyond calcium
channels? Curr Neuropharmacol 15(6):918–925
712 S. Mallik and P. C. Acharya
Jang SJ, Choi HW, Choi DL, Cho S, Rim HK, Choi HE, Kim KS, Huang M, Rhim H, Lee KT, Lee
JY (2013) In vitro cytotoxicity on human ovarian cancer cells by T-type calcium channel
blockers. Bioorg Med Chem Lett 23(24):6656–6662
Jangsangthong W, Kuzmenkina E, Böhnke AK, Herzig S (2011) Single-channel monitoring of
reversible L-type Ca2+ channel CaV α1-CaVβ subunit interaction. Biophys J 101
(11):2661–2670
Jarvis MF, Scott VE, McGaraughty S, Chu KL, Xu J, Niforatos W, Milicic I, Joshi S, Zhang Q, Xia
Z (2014) A peripherally acting, selective T-type calcium channel blocker, ABT-639, effectively
reduces nociceptive and neuropathic pain in rats. Biochem Pharmacol 89(4):536–544
Jin AH, Dutertre S, Kaas Q, Lavergne V, Kubala P, Lewis RJ, Alewood PF (2013) Transcriptomic
messiness in the venom duct of Conus miles contributes to conotoxin diversity. Mol Cell
Proteomics 12(12):3824–3833
Jokinen V (2017) Opioid analgesia: modulation by drug interactions and glial activation. PhD
thesis, University of Helsinki, Finland
Joksimovic SL, Joksimovic SM, Tesic V, García-Caballero A, Feseha S, Zamponi GW, Jevtovic-
Todorovic V, Todorovic SM (2018) Selective inhibition of CaV3. 2 channels reverses hyperex-
citability of peripheral nociceptors and alleviates postsurgical pain. Sci Signal 11(545):
eaao4425
Jurkovicova-Tarabova B, Lacinova L (2019) Structure, function and regulation of CaV 2.2 N-type
calcium channels. Gen Physiol Biophys. https://doi.org/10.4149/gpb_2019004
Kadurin I, Ferron L, Rothwell SW, Meyer JO, Douglas LR, Bauer CS, Lana B, Margas W,
Alexopoulos O, Nieto-Rostro M, Pratt WS (2016) Proteolytic maturation of α2δ represents a
checkpoint for activation and neuronal trafficking of latent calcium channels. Elife 5:e21143
Kang HW, Park JY, Jeong SW, Kim JA, Moon HJ, Perez-Reyes E, Lee JH (2006) A molecular
determinant of nickel inhibition in Cav3. 2 T-type calcium channels. J Biol Chem 281
(8):4823–4830
Kang S, Cooper G, Dunne SF, Dusel B, Luan CH, Surmeier DJ, Silverman RB (2012) CaV1.3-
selective L-type calcium channel antagonists as potential new therapeutics for Parkinson’s
disease. Nature Commun 3:1146
Kappel VD, Zanatta L, Postal BG, Silva FRMB (2013) Rutin potentiates calcium uptake via
voltage-dependent calcium channel associated with stimulation of glucose uptake in skeletal
muscle. Arch Biochem Biophys 532(2):55–60
Katzung BG (2017) Basic and clinical pharmacology. McGraw-Hill Education, New York, NY
Kazim AS (2017) Roles of voltage-gated Ca2+ channel subunits in pancreatic β cells. PhD thesis,
Lund University, Sweden
Kim SK, Kim TK (2018) New molecular insights into the excitation-transcription coupling. Neuron
98(3):453–456
Kim TY, Maki T, Zhou Y, Sakai K, Mizuno Y, Ishikawa A, Tanaka R, Niimi K, Li W, Nagano N,
Takahashi E (2015) Absence-like seizures and their pharmacological profile in tottering-6j mice.
Biochem Biophys Res Commun 463(1-2):148–153
King GF, Escoubas P, Nicholson GM (2008) Peptide toxins that selectively target insect NaV and
CaV channels. Channels 2(2):100–116
Klint JK, Senff S, Rupasinghe DB, Er SY, Herzig V, Nicholson GM, King GF (2012) Spider-
venom peptides that target voltage-gated sodium channels: pharmacological tools and potential
therapeutic leads. Toxicon 60(4):478–491
Korkosh VS, Kisilev AM, Mikhaylov EN, Kostareva AA, Zhorov BS (2019) Atomic mechanisms
of timothy syndrome-associated mutations in calcium channel Cav1. 2. Front Physiol 10:335
Krouse AJ, Gray L, Macdonald T, McCray J (2015) Repurposing and rescuing of mibefradil, an
antihypertensive, for cancer: a case study. Drug Repurposing Rescue Repositioning 1(1):36–39
Kuo IYT, Howitt L, Sandow SL, McFarlane A, Hansen PB, Hill CE (2014) Role of T-type channels
in vasomotor function: team player or chameleon? Pflügers Arch 466(4):767–779
Kuwahara K, Kimura T (2015) The organ-protective effect of N-type Ca2+ channel blockade.
Pharmacol Ther 151:1–7
20 Pharmacology of Calcium Channel 715
Kyle DJ, Park JH, Purdue Pharma LP (2017) Sodium channel blocking peptides and the use thereof.
US Patent 9,718,865
Larsch J, Flavell SW, Liu Q, Gordus A, Albrecht DR, Bargmann CI (2015) A circuit for gradient
climbing in C. elegans chemotaxis. Cell Rep 12(11):1748–1760
Latham JR, Pathirathna S, Jagodic MM, Choe WJ, Levin ME, Nelson MT, Lee WY, Krishnan K,
Covey DF, Todorovic SM, Jevtovic-Todorovic V (2009) Selective T-type calcium channel
blockade alleviates hyperalgesia in ob/ob mice. Diabetes 58(11):2656–2665
Laurent S (2017) Antihypertensive drugs. Pharmacol Res 124:116–125
Laurent B, Murail S, Shahsavar A, Sauguet L, Delarue M, Baaden M (2016) Sites of anesthetic
inhibitory action on a cationic ligand-gated ion channel. Structure 24(4):595–605
Lee AS, De Jesús-Cortés H, Kabir ZD, Knobbe W, Orr M, Burgdorf C, Huntington P, McDaniel L,
Britt JK, Hoffmann F, Brat DJ (2016) The neuropsychiatric disease-associated gene cacna1c
mediates survival of young hippocampal neurons. Eneuro 3(2). https://doi.org/10.1523/
ENEURO.0006-16.2016
Lee MS, Newbold EJ, Papapetropoulos S (2019) Selective T-type calcium channel modulator
CX-8998 fully suppresses seizures in the GAERS genetic model of epilepsy at human achiev-
able concentrations (P1. 5-026)
Lenaeus MJ, El-Din TMG, Ing C, Ramanadane K, Pomès R, Zheng N, Catterall WA (2017)
Structures of closed and open states of a voltage-gated sodium channel. Proc Natl Acad Sci
114(15):E3051–E3060
Lewis RJ, Nielsen KJ, Craik DJ, Loughnan ML, Adams DA, Sharpe IA, Luchian T, Adams DJ,
Bond T, Thomas L, Jones A (2000) Novel ω-conotoxins from Conus catus discriminate among
neuronal calcium channel subtypes. J Biol Chem 275(45):35335–35344
Li Y, Tatsui CE, Rhines LD, North RY, Harrison DS, Cassidy RM, Johansson CA, Kosturakis AK,
Edwards DD, Zhang H, Dougherty PM (2017a) Dorsal root ganglion neurons become hyperex-
citable and increase expression of voltage-gated T-type calcium channels (Cav3. 2) in
paclitaxel-induced peripheral neuropathy. Pain 158(3):417
Li G, Wang J, Liao P, Bartels P, Zhang H, Yu D, Liang MC, Poh KK, Yu CY, Jiang F, Yong TF
(2017b) Exclusion of alternative exon 33 of CaV1. 2 calcium channels in heart is
proarrhythmogenic. Proc Natl Acad Sci 114(21):E4288–E4295
Lian LY, Pandalaneni SR, Todd PA, Martin VM, Burgoyne RD, Haynes LP (2014) Demonstration
of binding of neuronal calcium sensor-1 to the cav2. 1 p/q-type calcium channel. Biochemistry
53(38):6052–6062
Liao P, Soong TW (2010) Ca V 1.2 channelopathies: from arrhythmias to autism, bipolar disorder,
and immunodeficiency. Pflügers Arch 460(2):353–359
Liu Z, Bartels P, Sadeghi M, Du T, Dai Q, Zhu C, Yu S, Wang S, Dong M, Sun T, Guo J (2018) A
novel α-conopeptide Eu1. 6 inhibits N-type (Ca V 2.2) calcium channels and exhibits potent
analgesic activity. Sci Rep 8(1):1004
Loperena R, Harrison DG (2017) Oxidative stress and hypertensive diseases. Med Clin 101
(1):169–193
Lotia M, Jankovic J (2016) New and emerging medical therapies in Parkinson’s disease. Expert
Opin Pharmacother 17(7):895–909
Lu L, Sirish P, Zhang Z, Woltz RL, Li N, Timofeyev V, Knowlton AA, Zhang XD, Yamoah EN,
Chiamvimonvat N (2015) Regulation of gene transcription by voltage-gated L-type calcium
channel, Cav1. 3. J Biol Chem 290(8):4663–4676
M’Dahoma S, Gadotti VM, Zhang FX, Park B, Nam JH, Onnis V, Balboni G, Lee JY, Zamponi
GW (2016) Effect of the T-type channel blocker KYS-05090S in mouse models of acute and
neuropathic pain. Pflügers Arch 468(2):193–199
Manthri S, Veena D, Ambari RB, Puchchakayala G (2015) A prospective interventional study on
clinical effects of cilnidipine in hypertensive patients. IJPRIF 8(10):70–76
Marchetti C (2013) Role of calcium channels in heavy metal toxicity. ISRN Toxicol 2013:184360
716 S. Mallik and P. C. Acharya
Mary R, Giribaldi J, Lesport P, Bourinet E, Dutertre S (2018) Discovery, synthesis and characteri-
zation of PmuTx1–A new spider toxin that blocks T-type calcium channels Cav3. 2. Toxicon
149:101
Matsunami M, Miki T, Nishiura K, Hayashi Y, Okawa Y, Nishikawa H, Sekiguchi F, Kubo L,
Ozaki T, Tsujiuchi T, Kawabata A (2012) Involvement of the endogenous hydrogen sulfide/
Cav3. 2 T-type Ca2+ channel pathway in cystitis-related bladder pain in mice. Br J Pharmacol
167(4):917–928
Matta C, Zákány R, Mobasheri A (2015) Voltage-dependent calcium channels in chondrocytes:
roles in health and disease. Curr Rheumatol Rep 17(7):43
McDonough SI (2007) Gating modifier toxins of voltage-gated calcium channels. Toxicon 49
(2):202–212
Medrihan L, Cesca F, Raimondi A, Lignani G, Baldelli P, Benfenati F (2013) Synapsin II
desynchronizes neurotransmitter release at inhibitory synapses by interacting with presynaptic
calcium channels. Nat Commun 4:1512
Mesirca P, Torrente AG, Mangoni ME (2015) Functional role of voltage gated Ca2+ channels in
heart automaticity. Front Physiol 6:19
Mir R, Karim S, Amjad Kamal M, Wilson MC, Mirza Z (2016) Conotoxins: structure, therapeutic
potential and pharmacological applications. Curr Pharm Des 22(5):582–589
Miriyala J, Nguyen T, Yue DT, Colecraft HM (2008) Role of CaVβ subunits, and lack of functional
reserve, in protein kinase A modulation of cardiac CaV1. 2 channels. Circ Res 102(7):e54–e64
Mollica A, Costante R, Novellino E, Stefanucci A, Pieretti S, Zador F, Samavati R, Borsodi A,
Benyhe S, Vetter I, Lewis RJ (2015) Design, synthesis and biological evaluation of two opioid
agonist and Cav2. 2 blocker multitarget ligands. Chem Biol Drug Des 86(2):156–162
Montandon G, Ren J, Victoria NC, Liu H, Wickman K, Greer JJ, Horner RL (2016) G-protein–
gated Inwardly Rectifying Potassium Channels Modulate Respiratory Depression by Opioids.
Anesthesiology 124(3):641–650
Morrill GA, Kostellow AB, Gupta RK (2015) Computational analysis of the extracellular domain of
the Ca2+-sensing receptor: an alternate model for the Ca2+ sensing region. Biochem Biophys
Res Commun 459(1):36–41
Motin L, Yasuda T, Schroeder CI, Lewis RJ, Adams DJ (2007) ω-Conotoxin CVIB differentially
inhibits native and recombinant N-and P/Q-type calcium channels. Eur J Neurosci 25
(2):435–444
Moutal A, Dustrude ET, Khanna R (2017) Sensitization of ion channels contributes to central and
peripheral dysfunction in neurofibromatosis type 1. Mol Neurobiol 54(5):3342–3349
Nam G (2018) T-type calcium channel blockers: a patent review (2012–2018). Expert Opin Ther
Pat 28(12):883–901
Nanclares C, Baraibar AM, Gandía L (2018) L-type calcium channels in exocytosis and endocytosis
of chromaffin cells. Pflügers Arch 470(1):53–60
Nanou E, Catterall WA (2018) Calcium channels, synaptic plasticity, and neuropsychiatric disease.
Neuron 98(3):466–481
Newman RA, Prehoda KE (2009) Intramolecular interactions between the Src homology
3 guanylate kinase domains of discs large regulate its function in asymmetric cell division. J
Biol Chem 284(19):12924–12932
Nieto-Rostro M, Ramgoolam K, Pratt WS, Kulik A, Dolphin AC (2018) Ablation of α2δ-1 inhibits
cell-surface trafficking of endogenous N-type calcium channels in the pain pathway in vivo.
Proc Natl Acad Sci 115(51):E12043–E12052
Nimmrich V, Eckert A (2013) Calcium channel blockers and dementia. Br J Pharmacol 169
(6):1203–1210
Norton RS (2017) Enhancing the therapeutic potential of peptide toxins. Expert Opin Drug
Discovery 12(6):611–623
Offord J, Isom LL (2016) Drugging the undruggable: gabapentin, pregabalin and the calcium
channel α2δ subunit. Crit Rev Biochem Mol Biol 51(4):246–256
20 Pharmacology of Calcium Channel 717
expression in soma of dorsal root ganglion neurons and peripheral axons of rat and mouse.
Neuroscience 250:263–274
Ross JA (2018) The role of norepinephrine in modulating amyloid beta peptides: implications for
neurodegenerative and psychiatric disease. Drexel University
Roullet JB, Spaetgens RL, Burlingame T, Feng ZP, Zamponi GW (1999) Modulation of neuronal
voltage-gated calcium channels by farnesol. J Biol Chem 274:25439–25446
Rousset M, Cens T, Menard C, Bowerman M, Bellis M, Brusés J, Raoul C, Scamps F, Charnet P
(2015) Regulation of neuronal high-voltage activated CaV2 Ca2+ channels by the small GTPase
RhoA. Neuropharmacology 97:201–209
Rui LI, Shen L, Huang Y (2016) Effect of gabapentin on Cav3. 2 channels in dorsal root ganglia of
rats with neuropathic pain. Chin J Anesthesiol 36(11):1371–1374
Sanchez-Sandoval AL, Gomora JC (2019) Contribution of voltage-gated sodium channel
β-subunits to cervical cancer cells metastatic behavior. Cancer Cell Int 19(1):35
Santi CM, Cayabyab FS, Sutton KG, McRory JE, Mezeyova J, Hamming KS, Parker D, Stea A,
Snutch TP (2002) Differential inhibition of T-type calcium channels by neuroleptics. J Neurosci
22(2):396–403
Santos R, Ursu O, Gaulton A, Bento AP, Donadi RS, Bologa CG, Karlsson A, Al-Lazikani B,
Hersey A, Oprea TI, Overington JP (2017) A comprehensive map of molecular drug targets. Nat
Rev Drug Discov 16(1):19
Santulli G, Lewis DR, Marks AR (2017) Physiology and pathophysiology of excitation–contraction
coupling: the functional role of ryanodine receptor. J Muscle Res Cell Motil 38(1):37–45
Schroeder CI, Doering CJ, Zamponi GW, Lewis RJ (2006) N-type calcium channel blockers: novel
therapeutics for the treatment of pain. Med Chem 2(5):535–543
Schwab A, Fabian A, Hanley PJ, Stock C (2012) Role of ion channels and transporters in cell
migration. Physiol Rev 92(4):1865–1913
Scott I, Ramirez-Reveco A, Parodi J (2016) The restraint of bovine sperm cell motility increases
survival: role of extracellular calcium in the phenomena. J Vet Sci Technol 7(359):2
Seo HN, Choi JY, Choe YJ, Kim Y, Rhim H, Lee SH, Kim J, Joo DJ, Lee JY (2007) Discovery of
potent T-type calcium channel blocker. Bioorg Med Chem Lett 17(21):5740–5743
Shetty R, Vivek G, Naha K, Tumkur A, Raj A, Bairy KL (2013) Excellent tolerance to cilnidipine in
hypertensives with amlodipine-induced edema. N Am J Med Sci 5(1):47
Siafis S, Tzachanis D, Samara M, Papazisis G (2018) Antipsychotic drugs: from receptor-binding
profiles to metabolic side effects. Curr Neuropharmacol 16(8):1210–1223
Silva RB, Greggio S, Venturin GT, da Costa JC, Gomez MV, Campos MM (2018) Beneficial
effects of the calcium channel blocker ctk 01512-2 in a mouse model of multiple sclerosis. Mol
Neurobiol 55(12):9307–9327
Simms BA, Zamponi GW (2014) Neuronal voltage-gated calcium channels: structure, function, and
dysfunction. Neuron 82(1):24–45
Snutch TP, Zamponi GW (2018) Recent advances in the development of T-type calcium channel
blockers for pain intervention. Br J Pharmacol 175(12):2375–2383
Song L, Espinoza-Fuenzalida IA, Etheridge S, Jones OT, Fitzgerald EM (2015) The R-domain:
identification of an N-terminal region of the α2δ-1 subunit which is necessary and sufficient for
its effects on Cav2. 2 calcium currents. Curr Mol Pharmacol 8(2):169–179
Spedding M, Paoletti R (1992) Classification of calcium channels and the sites of action of drugs
modifying channel function. Pharmacol Rev 44(3):363–376
Srebro D, Vuckovic S, Milovanovic A, Kosutic J, Savic Vujovic K, Prostran M (2017) Magnesium
in pain research: state of the art. Curr Med Chem 24(4):424–434
Stölting G, de Oliveira RC, Guzman RE, Miranda-Laferte E, Conrad R, Jordan N, Schmidt S,
Hendriks J, Gensch T, Hidalgo P (2015) Direct interaction of CaVβ with actin up-regulates
L-type calcium currents in HL-1 cardiomyocytes. J Biol Chem 290(8):4561–4572
Striessnig J, Pinggera A, Kaur G, Bock G, Tuluc P (2014) L-type Ca2+ channels in heart and brain.
Wiley Interdiscip Rev Membr Transp Signal 3(2):15–38
20 Pharmacology of Calcium Channel 719
Striessnig J, Ortner NJ, Pinggera A (2015) Pharmacology of L-type calcium channels: novel drugs
for old targets? Curr Mol Pharmacol 8(2):110–122
Südhof TC (2013) A molecular machine for neurotransmitter release: synaptotagmin and beyond.
Nat Med 19(10):1227
Surmeier DJ Jr, Silverman RB, Kang SS, Cooper G (2019) Northwestern University, 2019.
SELECTIVE CALCIUM CHANNEL ANTAGONISTS. U.S. Patent Application 16/167,055
Surmeier DJ, Schumacker PT, Guzman JD, Ilijic E, Yang B, Zampese E (2017) Calcium and
Parkinson’s disease. Biochem Biophys Res Commun 483(4):1013–1019
Szabó GG, Lenkey N, Holderith N, Andrási T, Nusser Z, Hájos N (2014) Presynaptic calcium
channel inhibition underlies CB1 cannabinoid receptor-mediated suppression of GABA release.
J Neurosci 34(23):7958–7963
Takahashi Y, Nishimura T, Higuchi K, Noguchi S, Tega Y, Kurosawa T, Deguchi Y, Tomi M
(2018) Transport of pregabalin via L-type amino acid transporter 1 (SLC7A5) in human brain
capillary endothelial cell line. Pharm Res 35(12):246
Tang L, El-Din TMG, Swanson TM, Pryde DC, Scheuer T, Zheng N, Catterall WA (2016)
Structural basis for inhibition of a voltage-gated Ca 2+ channel by Ca2+ antagonist drugs.
Nature 537(7618):117
Tano YK, Wakimoto S, Tamura S, Kubota K, Domon Y, Arakawa N, Saito M, Sava B, Buisson B
(2019) Effects of mirogabalin, a novel ligand for the α2δ subunit of voltage-gated calcium
channels, on N-type calcium channel currents of rat dorsal root ganglion culture neurons.
Pharmazie 74(3):147–149
Taylor CP (2009) Mechanisms of analgesia by gabapentin and pregabalin–Calcium channel α2-δ
[Cavα2-δ] ligands. Pain 142(1):13–16
Thalhammer A, Contestabile A, Ermolyuk YS, Ng T, Volynski KE, Soong TW, Goda Y, Cingolani
LA (2017) Alternative splicing of P/Q-Type Ca2+ channels shapes presynaptic plasticity. Cell
Rep 20(2):333–343
Thapa P, Espiritu MJ, Cabalteja CC, Bingham JP (2014) Conotoxins and their regulatory
considerations. Regul Toxicol Pharmacol 70(1):197–202
Thompson JC, Dunbar E, Laye RR (2006) Treatment challenges and complications with ziconotide
monotherapy in established pump patients. Pain Physician 9:147–152
Tikhonov DB, Zhorov BS (2017) Mechanism of sodium channel block by local anesthetics,
antiarrhythmics, and anticonvulsants. J Gen Physiol 149(4):465–481
Tomita S, Sekiguchi F, Deguchi T, Miyazaki T, Ikeda Y, Tsubota M, Yoshida S, Du Nguyen H,
Okada T, Toyooka N, Kawabata A (2019) Critical role of Cav3. 2 T-type calcium channels in
the peripheral neuropathy induced by bortezomib, a proteasome-inhibiting chemotherapeutic
agent, in mice. Toxicology 413:33–39
Torres M, Marcilla-Etxenike A, Fiol-deRoque MA, Escribá PV, Busquets X (2015) The unfolded
protein response in the therapeutic effect of hydroxy-DHA against Alzheimer’s disease. Apo-
ptosis 20(5):712–724
Tse G, Liu T, Li KHC, Laxton V, Wong AOT, Chan YWF, Keung W, Chan CW, Li RA (2017)
Tachycardia-bradycardia syndrome: electrophysiological mechanisms and future therapeutic
approaches. Int J Mol Med 39(3):519–526
Turnaturi R, Arico G, Ronsisvalle G, Pasquinucci L, Parenti C (2016) Multitarget opioid/non-
opioid ligands: a potential approach in pain management. Curr Med Chem 23(40):4506–4528
Uchitel OD, Di Guilmi MN, Urbano FJ, Gonzalez-Inchauspe C (2010) Acute modulation of
calcium currents and synaptic transmission by gabapentinoids. Channels 4(6):490–496
Uzieliene I, Bernotas P, Mobasheri A, Bernotiene E (2018) The role of physical stimuli on calcium
channels in chondrogenic differentiation of mesenchymal stem cells. Int J Mol Sci 19(10):2998
Vallejo-Illarramendi A, Toral-Ojeda I, Aldanondo G, de Munain AL (2014) Dysregulation of
calcium homeostasis in muscular dystrophies. Expert Rev Mol Med 16:e16
Vega-Vela NE, Osorio D, Avila-Rodriguez M, Gonzalez J, García-Segura LM, Echeverria V,
Barreto GE (2017) L-type calcium channels modulation by estradiol. Mol Neurobiol 54
(7):4996–5007
720 S. Mallik and P. C. Acharya
Voisin T, Bourinet E, Lory P (2016) Genetic alteration of the metal/redox modulation of Cav3.
2 T-type calcium channel reveals its role in neuronal excitability. J Physiol 594(13):3561–3574
Wagner S, Rokita AG, Anderson ME, Maier LS (2013) Redox regulation of sodium and calcium
handling. Antioxid Redox Signal 18(9):1063–1077
Wang F, Yan Z, Liu Z, Wang S, Wu Q, Yu S, Ding J, Dai Q (2016) Molecular basis of toxicity of
N-type calcium channel inhibitor MVIIA. Neuropharmacology 101:137–145
Wang AL, Iadecola C, Wang G (2017) New generations of dihydropyridines for treatment of
hypertension. J Geriatr Cardiol 14(1):67
Wang X, Chen Y, Meng Z, Zhang Q, Zhai J (2018) Effect of Trivalent “Calcium-like” Cations on
Ionic Transport Behaviors of Artificial Calcium-Responsive Nanochannels. J Phys Chem C 122
(43):24863–24870
Watanabe M, Ueda T, Shibata Y, Kumamoto N, Shimada S, Ugawa S (2015) Expression and
regulation of Cav3. 2 T-type calcium channels during inflammatory hyperalgesia in mouse
dorsal root ganglion neurons. PLoS One 10(5):e0127572
Weiss N, Zamponi GW (2017) Trafficking of neuronal calcium channels. Neuronal Signal
1(1):1–16
Winters BL, Christie MJ, Vaughan CW (2019) Electrophysiological actions of N/OFQ. Springer,
New York, NY
Wong FK, Nath AR, Chen RH, Gardezi SR, Li Q, Stanley EF (2014) Synaptic vesicle tethering and
the CaV2. 2 distal C-terminal. Front Cell Neurosci 8:71
Woo LA, Tkachenko S, Ding M, Plowright AT, Engkvist O, Andersson H, Drowley L, Barrett I,
Firth M, Akerblad P, Wolf MJ (2019) High-content phenotypic assay for proliferation of human
iPSC-derived cardiomyocytes identifies L-type calcium channels as targets. J Mol Cell Cardiol
127:204–214
Woon MT, Balijepalli RC (2015) L-type Ca2+ channel Cavb subunits associate with and differen-
tially regulate the cardiac Cav3. 2 T-type Ca2+ channel currents. Biophys J 108(2):578a–579a
Wright AB, Norimatsu Y, McIntosh JM, Elmslie KS (2015) Limited efficacy of α-conopeptides,
Vc1. 1 and RgIA, to inhibit sensory neuron CaV current. eNeuro 2(1). https://doi.org/10.1523/
ENEURO.0057-14.2015
Xu Y, Sun J, Liu H, Sun J, Yu Y, Su Y, Cui Y, Zhao M, Zhang J (2018) Scorpion toxins targeting
voltage-gated sodium channels associated with pain. Curr Pharm Biotechnol 19(11):848–855
Yang R, Xiong Z, Liu C, Liu L (2014a) Inhibitory effects of capsaicin on voltage-gated potassium
channels by TRPV1-independent pathway. Cell Mol Neurobiol 34(4):565–576
Yang YC, Tai CH, Pan MK, Kuo CC (2014b) The T-type calcium channel as a new therapeutic
target for Parkinson’s disease. Pflügers Arch 466(4):747–755
Yang S, Ben-Shalom R, Ahn M, Liptak AT, van Rijn RM, Whistler JL, Bender KJ (2016)
β-arrestin-dependent dopaminergic regulation of calcium channel activity in the axon initial
segment. Cell Rep 16(6):1518–1526
Yocum GT, Perez-Zoghbi JF, Danielsson J, Kuforiji AS, Zhang Y, Li G, Rashid Roni MS,
Kodali R, Stafford DC, Arnold LA, Cook JM (2018) A novel GABAA receptor ligand
MIDD0301 with limited blood-brain barrier penetration relaxes airway smooth muscle
ex vivo and in vivo. Am J Physiol Lung Cell Mol Physiol 316(2):L385–L390
Yoder N, Yoshioka C, Gouaux E (2018) Gating mechanisms of acid-sensing ion channels. Nature
555(7696):397
Zamponi GW (2016) Targeting voltage-gated calcium channels in neurological and psychiatric
diseases. Nat Rev Drug Discov 15(1):19
Zamponi GW, Diaz P (2017) UTI LP and University of Montana, 2017. T-type calcium channel
modulator and uses thereof. U.S. Patent Application 15/518,473
Zamponi GW, Lory P, Perez-Reyes E (2010) Role of voltage-gated calcium channels in epilepsy.
Pflügers Arch 460(2):395–403
Zamponi GW, Striessnig J, Koschak A, Dolphin AC (2015) The physiology, pathology, and
pharmacology of voltage-gated calcium channels and their future therapeutic potential.
Pharmacol Rev 67(4):821–870
20 Pharmacology of Calcium Channel 721
Zaydman MA, Kasimova MA, McFarland K, Beller Z, Hou P, Kinser HE, Liang H, Zhang G, Shi J,
Tarek M, Cui J (2014) Domain–domain interactions determine the gating, permeation, pharma-
cology, and subunit modulation of the IKs ion channel. Elife 3:e03606
Zhang S, Yang L, Zhang K, Liu X, Dai W, Zhang C, Yong Z, Li J, Zheng J (2015) ZC88, a novel
N-type calcium channel blocker from 4-amino-piperidine derivatives state-dependent inhibits
Cav2. 2 calcium channels. Brain Res 1605:12–21
Zhang Y, Cruickshanks N, Yuan F, Wang B, Pahuski M, Wulfkuhle J, Gallagher I, Koeppel AF,
Hatef S, Papanicolas C, Lee J (2017) Targetable T-type calcium channels drive glioblastoma.
Cancer Res 77(13):3479–3490
Zhao Y, Huang G, Wu J, Wu Q, Gao S, Yan Z, Lei J, Yan N (2019) Molecular basis for ligand
modulation of a mammalian voltage-gated Ca2+ channel. Cell 177(6):1495–1506
Zheng J (2013) Molecular mechanism of TRP channels. Compr Physiol 3(1):221–242
Zvejniece L, Vavers E, Svalbe B, Veinberg G, Rizhanova K, Liepins V, Kalvinsh I, Dambrova M
(2015) R-phenibut binds to the α2–δ subunit of voltage-dependent calcium channels and exerts
gabapentin-like anti-nociceptive effects. Pharmacol Biochem Behav 137:23–29