The Concise Guide To Pharmacology 201920 Transport
The Concise Guide To Pharmacology 201920 Transport
Abstract
The Concise Guide to PHARMACOLOGY 2019/20 is the fourth in this series of biennial publications. The Concise Guide provides concise overviews of the key properties of nearly 1800 human
drug targets with an emphasis on selective pharmacology (where available), plus links to the open access knowledgebase source of drug targets and their ligands (www.guidetopharmacology.org),
which provides more detailed views of target and ligand properties. Although the Concise Guide represents approximately 400 pages, the material presented is substantially reduced compared
to information and links presented on the website. It provides a permanent, citable, point-in-time record that will survive database updates. The full contents of this section can be found at
http://onlinelibrary.wiley.com/doi/10.1111/bph.14753. Transporters are one of the six major pharmacological targets into which the Guide is divided, with the others being: G protein-coupled re-
ceptors, ion channels, nuclear hormone receptors, catalytic receptors and enzymes. These are presented with nomenclature guidance and summary information on the best available pharmacological
tools, alongside key references and suggestions for further reading. The landscape format of the Concise Guide is designed to facilitate comparison of related targets from material contemporary to
mid-2019, and supersedes data presented in the 2017/18, 2015/16 and 2013/14 Concise Guides and previous Guides to Receptors and Channels. It is produced in close conjunction with the International
Union of Basic and Clinical Pharmacology Committee on Receptor Nomenclature and Drug Classification (NC-IUPHAR), therefore, providing official IUPHAR classification and nomenclature for human
drug targets, where appropriate.
Conflict of interest
The authors state that there are no conflicts of interest to disclose.
© 2019 The Authors. British Journal of Pharmacology published by John Wiley & Sons Ltd on behalf of The British Pharmacological Society.
This is an open access article under the terms of the Creative Commons Attribution License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.
Overview: The majority of biological solutes are charged organic teins, which transport (primarily) inorganic cations. The second, 400 members. Many of these overlap in terms of the solutes that
or inorganic molecules. Cellular membranes are hydrophobic F-type or V-type ATPases, are proton-coupled motors, which can they carry. For example, amino acids accumulation is mediated
and, therefore, effective barriers to separate them allowing the for- function either as transporters or as motors. Last, are ATP-binding by members of the SLC1, SLC3/7, SLC6, SLC15, SLC16, SLC17,
mation of gradients, which can be exploited, for example, in the cassette transporters, heavily involved in drug disposition as well SLC32, SLC36, SLC38 and SLC43 families. Further members of
generation of energy. Membrane transporters carry solutes across as transporting endogenous solutes. the SLC superfamily regulate ion fluxes at the plasma membrane,
cell membranes, which would otherwise be impermeable to them. The second largest family of membrane proteins in the human or solute transport into and out of cellular organelles. Some SLC
The energy required for active transport processes is obtained from genome, after the G protein-coupled receptors, are the SLC so- family members remain orphan transporters, in as much as a phys-
ATP turnover or by exploiting ion gradients. lute carrier family. Within the solute carrier family, there are a iological function has yet to be dtermined. Within the SLC super-
ATP-driven transporters can be divided into three major classes: P- great variety of solutes transported, from simple inorganic ions to family, there is an abundance in diversity of structure. Two fami-
type ATPases; F-type or V-type ATPases and ATP-binding cassette amino acids and sugars to relatively complex organic molecules lies (SLC3 and SLC7) only generate functional transporters as het-
transporters. The first of these, P-type ATPases, are multimeric pro- like haem. The solute carrier family includes 65 families of almost eromeric partners, where one partner is a single TM domain pro-
tein. Membrane topology predictions for other families suggest direction. Symports allow concentration gradients of one solute to dients. A more complex family of transporters, the SLC27 fatty
3,4,6,7,8,9,10,11,12,13 or 14 TM domains. The SLC transporters allow co-transport of a second solute across a membrane. A third, acid transporters also express enzymatic function. Many of the
include members which function as antiports, where solute move- relatively small group are equilibrative transporters, which allow transporters also express electrogenic properties of ion channels.
ment in one direction is balanced by a solute moving in the reverse solutes to travel across membranes down their concentration gra-
Family structure
S399 ATP-binding cassette transporter family S428 SLC8 family of sodium/calcium exchangers S457 SLC27 family of fatty acid transporters
S399 ABCA subfamily S429 SLC9 family of sodium/hydrogen exchangers S458 SLC28 and SLC29 families of nucleoside transporters
S401 ABCB subfamily S429 SLC10 family of sodium-bile acid co-transporters S458 SLC28 family
S403 ABCC subfamily S431 SLC11 family of proton-coupled metal ion transporters S459 SLC29 family
S404 ABCD subfamily of peroxisomal ABC transporters S431 SLC12 family of cation-coupled chloride transporters S461 SLC30 zinc transporter family
S405 ABCG subfamily S433 SLC13 family of sodium-dependent sulphate/carboxylate S461 SLC31 family of copper transporters
S406 F-type and V-type ATPases transporters S462 SLC32 vesicular inhibitory amino acid transporter
S406 F-type ATPase S434 SLC14 family of facilitative urea transporters S463 SLC33 acetylCoA transporter
S407 V-type ATPase S435 SLC15 family of peptide transporters S464 SLC34 family of sodium phosphate co-transporters
S407 P-type ATPases S437 SLC16 family of monocarboxylate transporters S465 SLC35 family of nucleotide sugar transporters
S407 Na+ /K+ -ATPases S438 SLC17 phosphate and organic anion transporter family S466 SLC36 family of proton-coupled amino acid transporters
S408 Ca2+ -ATPases S438 Type I sodium-phosphate co-transporters S468 SLC37 family of phosphosugar/phosphate exchangers
S408 H+ /K+ -ATPases S439 Sialic acid transporter S468 SLC38 family of sodium-dependent neutral
S408 Cu+ -ATPases S439 Vesicular glutamate transporters (VGLUTs) amino acid transporters
S409 Phospholipid-transporting ATPases S440 Vesicular nucleotide transporter S469 System A-like transporters
S409 SLC superfamily of solute carriers S440 SLC18 family of vesicular amine transporters S469 System N-like transporters
S410 SLC1 family of amino acid transporters S442 SLC19 family of vitamin transporters S470 Orphan SLC38 transporters
S410 Glutamate transporter subfamily S443 SLC20 family of sodium-dependent phosphate S470 SLC39 family of metal ion transporters
S412 Alanine/serine/cysteine transporter subfamily transporters S471 SLC40 iron transporter
S413 SLC2 family of hexose and sugar alcohol transporters S443 SLC22 family of organic cation and anion transporters S472 SLC41 family of divalent cation transporters
S413 Class I transporters S444 Organic cation transporters (OCT) S473 SLC42 family of Rhesus glycoprotein ammonium
S414 Class II transporters S445 Organic zwitterions/cation transporters (OCTN) transporters
S415 Proton-coupled inositol transporter S446 Organic anion transporters (OATs) S473 SLC43 family of large neutral amino acid transporters
S415 SLC3 and SLC7 families of heteromeric amino acid S446 Urate transporter S474 SLC44 choline transporter-like family
transporters (HATs) – Orphan or poorly characterized S475 SLC45 family of putative sugar transporters
S415 SLC3 family SLC22 family members S475 SLC46 family of folate transporters
S416 SLC7 family S447 Atypical SLC22B subfamily S477 SLC47 family of multidrug and toxin extrusion
S417 SLC4 family of bicarbonate transporters S448 SLC23 family of ascorbic acid transporters transporters
S417 Anion exchangers S449 SLC24 family of sodium/potassium/calcium exchangers S477 SLC48 heme transporter
S418 Sodium-dependent HCO3 - transporters S450 SLC25 family of mitochondrial transporters S478 SLC49 family of FLVCR-related heme transporters
S418 SLC5 family of sodium-dependent glucose transporters S450 Mitochondrial di- and tri-carboxylic acid S479 SLC50 sugar transporter
S419 Hexose transporter family transporter subfamily S479 SLC51 family of steroid-derived molecule transporters
S420 Choline transporter S451 Mitochondrial amino acid transporter subfamily S480 SLC52 family of riboflavin transporters
S421 Sodium iodide symporter, sodium-dependent S452 Mitochondrial phosphate transporters S481 SLC53 Phosphate carriers
multivitamin transporter and sodium-coupled S452 Mitochondrial nucleotide transporter subfamily S481 SLC54 Mitochondrial pyruvate carriers
monocarboxylate transporters S453 Mitochondrial uncoupling proteins S482 SLC55 Mitochondrial cation/proton exchangers
S422 Sodium myo-inositol cotransporter transporters S454 Miscellaneous SLC25 mitochondrial transporters S482 SLC56 Sideroflexins
S423 SLC6 neurotransmitter transporter family S454 SLC26 family of anion exchangers S483 SLC57 NiPA-like magnesium transporter family
S423 Monoamine transporter subfamily S454 Selective sulphate transporters S483 SLC58 MagT-like magnesium transporter family
S424 GABA transporter subfamily S455 Chloride/bicarbonate exchangers S484 SLC59 Sodium-dependent lysophosphatidylcholine
S425 Glycine transporter subfamily S455 Anion channels symporter family
S427 Neutral amino acid transporter subfamily S456 Other SLC26 anion exchangers S484 SLC60 Glucose transporters
S485 SLC61 Molybdate transporter family S486 SLC64 Golgi Ca2+ /H+ exchangers
S485 SLC62 Pyrophosphate transporters S487 SLC65 NPC-type cholesterol transporters
S486 SLC63 Sphingosine-phosphate transporters S488 SLCO family of organic anion transporting polypeptides
Overview: ATP-binding cassette transporters are ubiquitous (NBD). The majority of eukaryotic ABC transporters are ‘full’ trans- convey substrates from the cytoplasm, either out of the cell or into
membrane proteins characterized by active ATP-dependent move- porters incorporating both TM and NBD entities. Some ABCs, no- intracellular organelles. Their role in the efflux of exogenous com-
ment of a range of substrates, including ions, lipids, peptides, tably the ABCD and ABCG families are half-transporters with only pounds, notably chemotherapeutic agents, has led to considerable
steroids. Individual subunits are typically made up of two groups a single membrane spanning domain and one NBD, and are only interest.
of 6TM-spanning domains, with two nucleotide-binding domains functional as homo- or heterodimers. Eukaryotic ABC transporters
ABCA subfamily
Transporters → ATP-binding cassette transporter family → ABCA subfamily
Overview: To date, 12 members of the human ABCA subfamily are identified. They share a high degree of sequence conservation and have been mostly related with lipid trafficking in a wide range
of body locations. Mutations in some of these genes have been described to cause severe hereditary diseases related with lipid transport, such as fatal surfactant deficiency or harlequin ichthyosis. In
addition, most of them are hypothesized to participate in the subcellular sequestration of drugs, thereby being responsible for the resistance of several carcinoma cell lines against drug treatment [8].
Comments: A number of structural analogues are not found in man: Abca14 (ENSMUSG00000062017); Abca15 (ENSMUSG00000054746); Abca16 (ENSMUSG00000051900) and Abca17
(ENSMUSG00000035435).
ABCB subfamily
Transporters → ATP-binding cassette transporter family → ABCB subfamily
Overview: The ABCB subfamily is composed of four full transporters and two half transporters. This is the only human subfamily to have both half and full types of transporters. ABCB1 was discovered
as a protein overexpressed in certain drug resistant tumor cells. It is expressed primarily in the blood brain barrier and liver and is thought to be involved in protecting cells from toxins. Cells that
overexpress this protein exhibit multi-drug resistance [142].
ABCC subfamily
Transporters → ATP-binding cassette transporter family → ABCC subfamily
Overview: Subfamily ABCC contains thirteen members and nine of these transporters are referred to as Multidrug Resistance Proteins (MRPs). MRP proteins are found throughout nature and mediate
many important functions. They are known to be involved in ion transport, toxin secretion, and signal transduction [142].
Comments: ABCC7 (also known as CFTR, a 12TM ABC tion of the Guide. ABCC8 (ENSG00000006071, also known as lar K+ channels (Kir6.1-6.2), conferring nucleotide sensitivity to
transporter-type protein, is a cAMP-regulated epithelial cell mem- SUR1, sulfonylurea receptor 1) and ABCC9 (ENSG00000069431, these channels to generate the canonical KATP channels. ABCC13
brane Cl- channel involved in normal fluid transport across var- also known as SUR2, sulfonylurea receptor 2) are unusual in that (ENSG00000155288) is a possible pseudogene.
ious epithelia and can be viewed in the Chloride channels sec- they lack transport capacity but regulate the activity of particu-
Comments: ABCD4 (ENSG00000119688, also known as PMP69, PXMP1-L or P70R) is located at the lysosome and is involved in the transport of vitamin B12 (cobalamin) from lysosomes into the
cytosol [122].
ABCG subfamily
Transporters → ATP-binding cassette transporter family → ABCG subfamily
Overview: This family of ’half-transporters’ act as homo- or heterodimers; particularly ABCG5 and ABCG8 are thought to be obligate heterodimers. The ABCG5/ABCG heterodimer sterol transporter
structure has been determined [389], suggesting an extensive intracellular nucleotide binding domain linked to the transmembrane domains by a fold in the primary sequence. The functional ABCG2
transporter appears to be a homodimer with structural similarities to the ABCG5/ABCG8 heterodimer [609].
Comments on ATP-binding cassette transporter family: to be functional transporters, but appear to have a role in protein as ABC50 or TNF-α-stimulated ABC protein); ABCF2 (Q9UG63,
A further group of ABC transporter-like proteins have been iden- translation [110, 490]: ABCE1 (P61221, also known as OABP or 2’- also known as iron-inhibited ABC transporter 2) and ABCF3
tified to lack membrane spanning regions and are not believed 5’ oligoadenylate-binding protein); ABCF1 (Q8NE71, also known (Q9NUQ8).
Baker A et al. (2015) Peroxisomal ABC transporters: functions and mechanism. Biochem. Soc. Trans. López-Marqués RL et al. (2015) Structure and mechanism of ATP-dependent phospholipid trans-
43: 959-65 [PMID:26517910] porters. Biochim. Biophys. Acta 1850: 461-475 [PMID:24746984]
Beis K. (2015) Structural basis for the mechanism of ABC transporters. Biochem. Soc. Trans. 43: Neul C et al. (2016) Impact of Membrane Drug Transporters on Resistance to Small-Molecule Tyrosine
889-93 [PMID:26517899] Kinase Inhibitors. Trends Pharmacol. Sci. 37: 904-932 [PMID:27659854]
Chen Z et al. (2016) Mammalian drug efflux transporters of the ATP binding cassette (ABC) family
in multidrug resistance: A review of the past decade. Cancer Lett. 370: 153-64 [PMID:26499806] Peña-Solórzano D et al. (2017) ABCG2/BCRP: Specific and Nonspecific Modulators. Med Res Rev 37:
Kemp S et al. (2011) Mammalian peroxisomal ABC transporters: from endogenous substrates to 987-1050 [PMID:28005280]
pathology and clinical significance. Br. J. Pharmacol. 164: 1753-66 [PMID:21488864] Robey RW et al. (2018) Revisiting the role of ABC transporters in multidrug-resistant cancer. Nat Rev
Kerr ID et al. (2011) The ABCG family of membrane-associated transporters: you don’t have to be Cancer 18: 452-464 [https://www.ncbi.nlm.nih.gov/pubmed/29643473]
big to be mighty. Br. J. Pharmacol. 164: 1767-79 [PMID:21175590] Vauthier V et al. (2017) Targeted pharmacotherapies for defective ABC transporters. Biochem.
Kloudova A et al. (2017) The Role of Oxysterols in Human Cancer. Trends Endocrinol. Metab. 28: Pharmacol. 136: 1-11 [PMID:28245962]
485-496 [PMID:28410994]
Overview: The F-type (ATP synthase) and the V-type (vacuolar complex (termed F1 or V1 ) and a membrane complex (Fo or Vo ). type and V-type ATPases have been assigned enzyme commission
or vesicular proton pump) ATPases, although having distinct sub- Within each ATPase complex, the two individual sectors appear to number E.C. 3.6.3.14
cellular locations and roles, exhibit marked similarities in subunit function as connected opposing rotary motors, coupling catalysis
structure and mechanism. They are both composed of a ‘soluble’ of ATP synthesis or hydrolysis to proton transport. Both the F-
F-type ATPase
Transporters → F-type and V-type ATPases → F-type ATPase
Overview: The F-type ATPase, also known as ATP synthase although it is also possible for the enzyme to function as an AT- The F0 motor, responsible for ion translocation, is complex in
or ATP phosphohydrolase (H+ -transporting), is a mitochondrial Pase. The ATP5O subunit (oligomycin sensitivity-conferring pro- mammals, with probably nine subunits centring on A, B, and C
membrane-associated multimeric complex consisting of two do- tein, OSCP, (P48047)), acts as a connector between F1 and F0 mo- subunits in the membrane, together with D, E, F2, F6, G2 and 8
mains, an F0 channel domain in the membrane and an F1 do- tors. subunits. Multiple pseudogenes for the F0 motor proteins have
main extending into the lumen. Proton transport across the in- The F1 motor, responsible for ATP turnover, has the subunit com- been defined in the human genome.
ner mitochondrial membrane is used to drive the synthesis of ATP, position α3β3γδ.
V-type ATPase
Transporters → F-type and V-type ATPases → V-type ATPase
Overview: The V-type ATPase is most prominently associated with lysosomes in mammals, but also appears to be expressed on the plasma membrane and neuronal synaptic vesicles.
The V1 motor, responsible for ATP turnover, has eight subunits with a composition of A-H.
TheV0 motor, responsible for ion translocation, has six subunits (a-e).
Brandt K et al. (2015) Hybrid rotors in F1F(o) ATP synthases: subunit composition, distribution, and Noji H et al. (2017) Catalytic robustness and torque generation of the F_1-ATPase. Biophys Rev 9:
physiological significance. Biol. Chem. 396: 1031-42 [PMID:25838297] 103-118 [PMID:28424741]
Krah A. (2015) Linking structural features from mitochondrial and bacterial F-type ATP synthases Okuno D et al. (2013) Single-molecule analysis of the rotation of F_1-ATPase under high hydrostatic
to their distinct mechanisms of ATPase inhibition. Prog. Biophys. Mol. Biol. 119: 94-102 pressure. Biophys. J. 105: 1635-42 [PMID:24094404]
[PMID:26140992]
Marshansky V et al. (2014) Eukaryotic V-ATPase: novel structural findings and functional insights.
Biochim. Biophys. Acta 1837: 857-79 [PMID:24508215]
P-type ATPases
Transporters → P-type ATPases
Overview: Phosphorylation-type ATPases (EC 3.6.3.-) are associ- between E1 and E2 conformations in the activity cycle of the trans- five subfamilies, P1-P5. The P1 subfamily includes heavy metal
ated with membranes and the transport of ions or phospholipids. porters, taken to represent ‘half-channels’ facing the cytoplasm pumps, such as the copper ATPases. The P2 subfamily includes
Characteristics of the family are the transient phosphorylation of and extracellular/luminal side of the membrane, respectively. calcium, sodium/potassium and proton/potassium pumps. The
the transporters at an aspartate residue and the interconversion Sequence analysis across multiple species allows the definition of P4 and P5 subfamilies include putative phospholipid flippases.
Na+/K+-ATPases
Transporters → P-type ATPases → Na+ /K+ -ATPases
Overview: The cell-surface Na+ /K+ -ATPase is an integral mem- hydrolysed, the Na+ /K+ -ATPase extrudes three Na+ ions and im- glycoprotein β-subunits. Additional protein partners known as
brane protein which regulates the membrane potential of the cell ports two K+ ions. The active transporter is a heteromultimer with FXYD proteins (e.g. FXYD2, P54710) appear to associate with and
by maintaining gradients of Na+ and K+ ions across the plasma incompletely defined stoichiometry, possibly as tetramers of het- regulate the activity of the pump.
membrane, also making a small, direct contribution to membrane erodimers, each consisting of one of four large, ten TM domain
potential, particularly in cardiac cells. For every molecule of ATP catalytic α subunits and one of three smaller, single TM domain
Comments: Na+ /K+ -ATPases are inhibited by ouabain and cardiac glycosides, such as digoxin, as well as potentially endogenous cardiotonic steroids [34].
Ca2+-ATPases
Transporters → P-type ATPases → Ca2+ -ATPases
Overview: The sarcoplasmic/endoplasmic reticulum Ca2+ - The plasma membrane Ca2+ -ATPase (PMCA) is a cell-surface pump loops.
ATPase (SERCA) is an intracellular membrane-associated pump for extruding calcium from the cytosol, usually associated with the Secretory pathway Ca2+ -ATPases (SPCA) allow accumulation of
for sequestering calcium from the cytosol into intracellular or- recovery phase following excitation of cells. The active pump is calcium and manganese in the Golgi apparatus.
ganelles, usually associated with the recovery phase following ex- a homodimer, each subunit of which is made up of ten TM seg-
citation of muscle and nerves. ments, with cytosolic C- and N-termini and two large intracellular
Comments: The fungal toxin ochratoxin A has been described to activate SERCA in kidney microsomes [116]. Cyclopiazonic acid [564], thapsigargin [414] and BHQ are widely employed to block SERCA.
Thapsigargin has also been described to block the TRPV1 vanilloid receptor [629].
The stoichiometry of flux through the PMCA differs from SERCA, with the PMCA transporting 1 Ca2+ while SERCA transports 2 Ca2+ .
Loss-of-function mutations in SPCA1 appear to underlie Hailey-Hailey disease [299].
H+/K+-ATPases
Transporters → P-type ATPases → H+ /K+ -ATPases
Overview: The H+ /K+ ATPase is a heterodimeric protein, made up of α and β subunits. The α subunit has 10 TM domains and exhibits catalytic and pore functions, while the β subunit has a single
TM domain, which appears to be required for intracellular trafficking and stabilising the α subunit. The ATP4A and ATP4B subunits are expressed together, while the ATP12A subunit is suggested to be
expressed with the β1 (ATP1B1) subunit of the Na+ /K+ -ATPase [495].
Comments: The gastric H+ /K+ -ATPase is inhibited by proton pump inhibitors used for treating excessive gastric acid secretion, including dexlansoprazole and a metabolite of esomeprazole.
Cu+-ATPases
Transporters → P-type ATPases → Cu+ -ATPases
Overview: Copper-transporting ATPases convey copper ions across cell-surface and intracellular membranes. They consist of eight TM domains and associate with multiple copper chaperone proteins
(e.g. ATOX1, O00244).
Phospholipid-transporting ATPases
Transporters → P-type ATPases → Phospholipid-transporting ATPases
Overview: These transporters are thought to translocate the aminophospholipids phosphatidylserine and phosphatidylethanolamine from one side of the phospholipid bilayer to the other to generate
asymmetric membranes. They are also proposed to be involved in the generation of vesicles from intracellular and cell-surface membranes.
Comments: Loss-of-function mutations in ATP8B1 are associated A further series of structurally-related proteins have been iden- including ATP13A1 (Q9HD20), ATP13A2 (Q9NQ11), ATP13A3
with type I familial intrahepatic cholestasis. tified in the human genome, with as yet undefined function, (Q9H7F0), ATP13A4 (Q4VNC1) and ATP13A5 (Q4VNC0).
Aperia A et al. (2016) Na+-K+-ATPase, a new class of plasma membrane receptors. Am. J. Physiol., Cell López-Marqués RL et al. (2015) Structure and mechanism of ATP-dependent phospholipid trans-
Physiol. 310: C491-5 [PMID:26791490] porters. Biochim. Biophys. Acta 1850: 461-475 [PMID:24746984]
Brini M et al. (2017) The plasma membrane calcium pumps: focus on the role in (neuro)pathology. Migocka M. (2015) Copper-transporting ATPases: The evolutionarily conserved machineries for bal-
Biochem. Biophys. Res. Commun. 483: 1116-1124 [PMID:27480928] ancing copper in living systems. IUBMB Life 67: 737-45 [PMID:26422816]
Bruce JIE. (2018) Metabolic regulation of the PMCA: Role in cell death and survival. Cell Calcium 69:
28-36 [PMID:28625348] Padányi R et al. (2016) Multifaceted plasma membrane Ca(2+) pumps: From structure to intracellular
Diederich M et al. (2017) Cardiac glycosides: From molecular targets to immunogenic cell death. Ca(2+) handling and cancer. Biochim. Biophys. Acta 1863: 1351-63 [PMID:26707182]
Biochem. Pharmacol. 125: 1-11 [PMID:27553475] Pomorski TG et al. (2016) Lipid somersaults: Uncovering the mechanisms of protein-mediated lipid
Dubois C et al. (2016) The calcium-signaling toolkit: Updates needed. Biochim. Biophys. Acta 1863: flipping. Prog. Lipid Res. 64: 69-84 [PMID:27528189]
1337-43 [PMID:26658643]
Krebs J. (2015) The plethora of PMCA isoforms: Alternative splicing and differential expression. Retamales-Ortega R et al. (2016) P2C-Type ATPases and Their Regulation. Mol. Neurobiol. 53: 1343-54
Biochim. Biophys. Acta 1853: 2018-24 [PMID:25535949] [PMID:25631710]
Little R et al. (2016) Plasma membrane calcium ATPases (PMCAs) as potential targets for the treat- Tadini-Buoninsegni F et al. (2017) Mechanisms of charge transfer in human copper ATPases ATP7A
ment of essential hypertension. Pharmacol. Ther. 159: 23-34 [PMID:26820758] and ATP7B. IUBMB Life 69: 218-225 [PMID:28164426]
Overview: The SLC superfamily of solute carriers is the second SLC1, SLC3/7, SLC6, SLC15, SLC16, SLC17, SLC32, SLC36, SLC38 Functionally, members may be divided into those dependent on
largest family of membrane proteins after G protein-coupled re- and SLC43. Further members of the SLC superfamily regulate ion gradients of ions (particularly sodium, chloride or protons), ex-
ceptors, but with a great deal fewer therapeutic drugs that exploit fluxes at the plasma membrane, or solute transport into and out change of solutes or simple equilibrative gating. For many mem-
them. As with the ABC transporters, however, they play a major of cellular organelles. bers, the stoichiometry of transport is not yet established. Fur-
role in drug disposition and so can be hugely influential in deter- Within the SLC superfamily, there is an abundance in diversity of thermore, one family of transporters also possess enzymatic activ-
mining the clinical efficacy of particular drugs. structure. Two families (SLC3 and SLC7) only generate functional ity (SLC27), while many members function as ion channels (e.g.
48 families are identified on the basis of sequence similarities, but transporters as heteromeric partners, where one partner is a sin- SLC1A7/EAAT5), which increases the complexity of function of
many of them overlap in terms of the solutes that they carry. For gle TM domain protein. Membrane topology predictions for other the SLC superfamily.
example, amino acid accumulation is mediated by members of the families suggest 3, 4 6, 7, 8, 9, 10, 11, 12, 13, or 14 TM domains.
Overview: Glutamate transporters present the unusual structural low ambient extracellular concentrations of glutamate (protecting moter activation [226, 390, 536] reviewed by [353]). PPARγ acti-
motif of 8TM segments and 2 re-entrant loops [262]. The crystal against excitotoxicity) and provide glutamate for metabolism in- vation (e.g. by rosiglitazone) also leads to enhanced expression
structure of a glutamate transporter homologue (GltPh) from Py- cluding the glutamate-glutamine cycle. The Na+ /K+ -ATPase that of EAAT though promoter activation [532]. In addition, several
rococcus horikoshii supports this topology and indicates that the maintains the ion gradients that drive transport has been demon- translational activators of EAAT2 have recently been described
transporter assembles as a trimer, where each monomer is a func- strated to co-assemble with EAAT1 and EAAT2 [533]. Recent ev- [125] along with treatments that increase the surface expression of
tional unit capable of substrate permeation [68, 526, 696] re- idence supports altered glutamate transport and novel roles in EAAT2 (e.g. [384, 726]), or prevent its down-regulation (e.g. [248]).
viewed by [329]). This structural data is in agreement with the pro- brain for splice variants of EAAT1 and EAAT2 [231, 386]. Three pa-
A thermodynamically uncoupled Cl- flux, activated by Na+ and
posed quaternary structure for EAAT2 [232] and several functional tients with dicarboxylic aminoaciduria (DA) were recently found
studies that propose the monomer is the functional unit [257, 366, to have loss-of-function mutations in EAAT3 [35]. DA is charac- glutamate [259, 339, 419] (Na+ and aspartate in the case of GltPh
385, 539]. Recent evidence suggests that EAAT3 and EAAT4 may terized by excessive excretion of the acidic amino acids glutamate [538]), is sufficiently large, in the instances of EAAT4 and EAAT5,
assemble as heterotrimers [467]. The activity of glutamate trans- and aspartate and EAAT3 is the predominant glutamate/aspartate to influence neuronal excitability [621, 648]. Indeed, it has re-
porters located upon both neurones (predominantly EAAT3, 4 and transporter in the kidney. Enhanced expression of EAAT2 result- cently been suggested that the primary function of EAAT5 is as a
5) and glia (predominantly EAAT 1 and 2) serves, dependent upon ing from administration of β-lactam antibiotics (e.g. ceftriaxone) is slow anion channel gated by glutamate, rather than a glutamate
their location, to regulate excitatory neurotransmission, maintain neuroprotective and occurs through NF-κB-mediated EAAT2 pro- transporter [220].
Nomenclature Excitatory amino acid transporter 1 Excitatory amino acid transporter 2 Excitatory amino acid transporter 3 Excitatory amino acid transporter 4 Excitatory amino acid transporter 5
Systematic nomenclature SLC1A3 SLC1A2 SLC1A1 SLC1A6 SLC1A7
Common abbreviation EAAT1 EAAT2 EAAT3 EAAT4 EAAT5
HGNC, UniProt SLC1A3, P43003 SLC1A2, P43004 SLC1A1, P43005 SLC1A6, P48664 SLC1A7, O00341
Substrates DL-threo-β-hydroxyaspartate (Ki D-aspartic acid, L-trans-2,4-pyrolidine D-aspartic acid, D-aspartic acid,
5.8×10−5 M) [571], D-aspartic acid, DL-threo-β-hydroxyaspartate, dicarboxylate, DL-threo-β-hydroxyaspartate, L-trans-2,4-pyrolidine
L-trans-2,4-pyrolidine L-trans-2,4-pyrolidine DL-threo-β-hydroxyaspartate, L-trans-2,4-pyrolidine dicarboxylate,
dicarboxylate dicarboxylate [367] D-aspartic acid dicarboxylate DL-threo-β-hydroxyaspartate
Endogenous substrates L-aspartic acid, L-glutamic acid L-glutamic acid, L-aspartic acid L-aspartic acid, L-cysteine [706], L-glutamic acid, L-aspartic acid L-aspartic acid, L-glutamic acid
L-glutamic acid
Stoichiometry Probably 3 Na+ : 1 H+ : 1 3 Na+ : 1 H+ : 1 glutamate (in): 1 3 Na+ : 1 H+ : 1 glutamate (in): 1 Probably 3 Na+ : 1 H+ : 1 Probably 3 Na+ : 1 H+ : 1
glutamate (in): 1 K+ (out) K+ (out) [396] K+ (out) [705] glutamate (in): 1 K+ (out) glutamate (in): 1 K+ (out)
Inhibitors UCPH-101 (membrane potential WAY-213613 (pIC50 7.1) [167], NBI-59159 (pIC50 7.1) [165], DL-TBOA (pKi 5.4) [570], DL-TBOA (pKi 5.5) [570]
assay) (pIC50 6.9) [326], DL-TBOA DL-TBOA (pKB 6.9) [571], L-β-BA ([3 H]D-aspartate uptake threo-3-methylglutamate (pKi 4.3)
(pKB 5) [571] SYM2081 (pKB 5.5) [640], assay) (pKi 6.1) [186], DL-TBOA [176]
dihydrokainate (pKB 5), (pIC50 5.1) [573]
threo-3-methylglutamate (pKB
4.7) [640]
Labelled ligands [3 H]ETB-TBOA (Binding) (pKd 7.8) [3 H]ETB-TBOA (Binding) (pKd 7.8) [3 H]ETB-TBOA (Binding) (pKd 6.5) [3 H]ETB-TBOA (Binding) (pKd 7.9) [3 H]ETB-TBOA (Binding) (pKd 7.6)
[572] – Rat, [3 H]D-aspartic acid, [572] – Rat, [3 H]D-aspartic acid, [572] – Rat, [3 H]D-aspartic acid, [572] – Rat, [3 H]D-aspartic acid, [572] – Rat, [3 H]D-aspartic acid,
[3 H]L-aspartic acid, [3 H]SYM2081 [3 H]L-aspartic acid, [3 H]SYM2081 [3 H]L-aspartic acid [3 H]L-aspartic acid [3 H]L-aspartic acid
Comments: The KB (or Ki ) values reported, unless indicated oth- ence for EAAT2 over EAAT3 and EAAT1 [166, 167]. NBI-59159 substrate-like currents at EAAT4, but does not elicit heteroex-
erwise, are derived from transporter currents mediated by EAATs is a non-substrate inhibitor with modest selectivity for EAAT3 change of [3 H]-aspartate in synaptosome preparations, inconsis-
expressed in voltage-clamped Xenopus laevis oocytes [176, 570, over EAAT1 (>10-fold) and EAAT2 (5-fold) [126, 164]. Analo- tent with the behaviour of a substrate inhibitor [176]. Parawixin 1,
571, 640]. KB (or Ki ) values derived in uptake assays are generally gously, L-β-threo-benzyl-aspartate (L-β-BA) is a competitive non- a compound isolated from the venom from the spider Parawixia
higher (e.g. [571]). In addition to acting as a poorly transportable substrate inhibitor that preferentially blocks EAAT3 versus EAAT1, bistriata is a selective enhancer of the glutamate uptake through
inhibitor of EAAT2, (2S,4R)-4-methylglutamate, also known as or EAAT2 [186]. [3 H]SYM2081 demonstrates low affinity binding EAAT2 but not through EAAT1 or EAAT3 [207, 208]. In addition
SYM2081, is a competitive substrate for EAAT1 (KM = 54μM; [300, (KD ∼= 6.0 μM) to EAAT1 and EAAT2 in rat brain homogenates to the agents listed in the table, DL-threo-β-hydroxyaspartate and
640]) and additionally is a potent kainate receptor agonist [715] [25] and EAAT1 in murine astrocyte membranes [23], whereas L-trans-2,4-pyrolidine dicarboxylate act as non-selective compet-
which renders the compound unsuitable for autoradiographic lo- [3 H]ETB-TBOA binds with high affinity to all EAATs other than itive substrate inhibitors of all EAATs. Zn2+ and arachidonic acid
calisation of EAATs [24]. Similarly, at concentrations that inhibit EAAT3 [572]. The novel isoxazole derivative (-)-HIP-A may in- are putative endogenous modulators of EAATs with actions that
EAAT2, dihydrokainate binds to kainate receptors [571]. WAY-855 teract at the same site as TBOA and preferentially inhibit reverse differ across transporter subtypes (reviewed by [639]).
and WAY-213613 are both non-substrate inhibitors with a prefer- transport of glutamate [124]. Threo-3-methylglutamate induces
Comments: The substrate specificity of ASCT1 may extend to L-proline and trans-4-hydroxy-proline [499]. At low pH (5.5) both ASCT1 and ASCT2 are able to exchange acidic amino acids such as
L-cysteate and glutamate [605, 634]. In addition to the inhibitors tabulated above, HgCl2 , methylmercury and mersalyl, at low micromolar concentrations, non-competitively inhibit ASCT2 by covalent
modificiation of cysteine residues [480].
Beart PM et al. (2007) Transporters for L-glutamate: an update on their molecular pharmacology and Jensen AA et al. (2015) Excitatory amino acid transporters: recent insights into molecular mecha-
pathological involvement. Br. J. Pharmacol. 150: 5-17 [PMID:17088867] nisms, novel modes of modulation and new therapeutic possibilities. Curr Opin Pharmacol 20:
Bjrn-Yoshimoto WE et al. (2016) The importance of the excitatory amino acid transporter 3 (EAAT3). 116-23 [PMID:25466154]
Neurochem. Int. 98: 4-18 [PMID:27233497] Kanai Y et al. (2013) The SLC1 high-affinity glutamate and neutral amino acid transporter family.
Fahlke C et al. (2016) Molecular physiology of EAAT anion channels. Pflugers Arch. 468: 491-502 Mol. Aspects Med. 34: 108-20 [PMID:23506861]
[PMID:26687113] Takahashi K et al. (2015) Glutamate transporter EAAT2: regulation, function, and potential as a
Fontana AC. (2015) Current approaches to enhance glutamate transporter function and expression.
therapeutic target for neurological and psychiatric disease. Cell. Mol. Life Sci. 72: 3489-506
J. Neurochem. 134: 982-1007 [PMID:26096891]
[PMID:26033496]
Grewer C et al. (2014) SLC1 glutamate transporters. Pflugers Arch. 466: 3-24 [PMID:24240778]
Class I transporters
Transporters → SLC superfamily of solute carriers → SLC2 family of hexose and sugar alcohol transporters → Class I transporters
Overview: Class I transporters are able to transport D-glucose, but not D-fructose, in the direction of the concentration gradient and may be inhibited non-selectively by phloretin and cytochalasin B.
GLUT1 is the major glucose transporter in brain, placenta and erythrocytes, GLUT2 is found in the pancreas, liver and kidneys, GLUT3 is neuronal and placental, while GLUT4 is the insulin-responsive
transporter found in skeletal muscle, heart and adipose tissue. GLUT14 appears to result from gene duplication of GLUT3 and is expressed in the testes [678].
Nomenclature Glucose transporter 1 Glucose transporter 2 Glucose transporter 3 Glucose transporter 4 Glucose transporter 14
Systematic nomenclature SLC2A1 SLC2A2 SLC2A3 SLC2A4 SLC2A14
Common abbreviation GLUT1 GLUT2 GLUT3 GLUT4 GLUT14
HGNC, UniProt SLC2A1, P11166 SLC2A2, P11168 SLC2A3, P11169 SLC2A4, P14672 SLC2A14, Q8TDB8
Substrates D-glucosamine (D-glucose = D-glucosamine (D-glucosamine > D-glucose D-glucosamine (D-glucosamine ≥ –
D-glucosamine) [631], D-glucose) [631], D-glucose D-glucose) [631], D-glucose
dehydroascorbic acid [55], D-glucose (D-glucosamine > D-glucose) [631] (D-glucosamine ≥ D-glucose) [631]
(D-glucose = D-glucosamine) [631]
Labelled ligands [3 H]2-deoxyglucose [3 H]2-deoxyglucose [3 H]2-deoxyglucose [3 H]2-deoxyglucose –
Comments GLUT1 is a class I facilitative sugar – – – –
transporter. GLUT1 functions to
maintain basal glucose import which
is required for cellular respiration.
Class II transporters
Transporters → SLC superfamily of solute carriers → SLC2 family of hexose and sugar alcohol transporters → Class II transporters
Overview: Class II transporters transport D-fructose and appear to be insensitive to cytochalasin B. Class II transporters appear to be predominantly intracellularly located.
Nomenclature Glucose transporter 11 Glucose transporter 6 Glucose transporter 8 Glucose transporter 10 Glucose transporter 12
Systematic nomenclature SLC2A11 SLC2A6 SLC2A8 SLC2A10 SLC2A12
Common abbreviation GLUT11 GLUT6 GLUT8 GLUT10 GLUT12
HGNC, UniProt SLC2A11, Q9BYW1 SLC2A6, Q9UGQ3 SLC2A8, Q9NY64 SLC2A10, O95528 SLC2A12, Q8TD20
Substrates D-fructose [426], D-glucose [156] – D-glucose [303] dehydroascorbic acid [392], D-glucose [530]
D-glucose [392]
Augustin R. (2010) The protein family of glucose transport facilitators: It’s not only about glucose Mueckler M et al. (2013) The SLC2 (GLUT) family of membrane transporters. Mol. Aspects Med. 34:
after all. IUBMB Life 62: 315-33 [PMID:20209635] 121-38 [PMID:23506862]
Klip A et al. (2014) Signal transduction meets vesicle traffic: the software and hardware of GLUT4 Uldry M et al. (2004) The SLC2 family of facilitated hexose and polyol transporters. Pflugers Arch.
translocation. Am. J. Physiol., Cell Physiol. 306: C879-86 [PMID:24598362] 447: 480-9 [PMID:12750891]
Leney SE et al. (2009) The molecular basis of insulin-stimulated glucose uptake: signalling, trafficking
and potential drug targets. J. Endocrinol. 203: 1-18 [PMID:19389739]
SLC3 family
Transporters → SLC superfamily of solute carriers → SLC3 and SLC7 families of heteromeric amino acid transporters (HATs) → SLC3 family
Overview: SLC3 family members are single TM proteins with extensive glycosylation of the exterior C-terminus, which heterodimerize with SLC7 family members in the endoplasmic reticulum and
assist in the plasma membrane localization of the transporter.
SLC7 family
Transporters → SLC superfamily of solute carriers → SLC3 and SLC7 families of heteromeric amino acid transporters (HATs) → SLC7 family
Overview: SLC7 family members may be divided into two major groups: cationic amino acid transporters (CATs) and glycoprotein-associated amino acid transporters (gpaATs).
Cationic amino acid transporters are 14 TM proteins, which mediate pH- and sodium-independent transport of cationic amino acids (system y+ ), apparently as an exchange mechanism. These transporters
are sensitive to inhibition by N-ethylmaleimide.
Nomenclature High affinity cationic amino acid Low affinity cationic amino acid Cationic amino acid transporter 3 L-type amino acid transporter 1 L-type amino acid transporter 2
transporter 1 transporter 2
Systematic nomenclature SLC7A1 SLC7A2 SLC7A3 SLC7A5 SLC7A8
Common abbreviation CAT1 CAT2 CAT3 LAT1 LAT2
HGNC, UniProt SLC7A1, P30825 SLC7A2, P52569 SLC7A3, Q8WY07 SLC7A5, Q01650 SLC7A8, Q9UHI5
Substrates L-ornithine, L-arginine, L-lysine, L-ornithine, L-arginine, L-lysine, L-ornithine, L-arginine, L-lysine – –
L-histidine L-histidine
Selective inhibitors – – – KYT-0353 [476] –
Nomenclature y+L amino acid transporter 1 y+L amino acid transporter 2 b0,+ -type amino acid Asc-type amino acid Cystine/glutamate transporter AGT1
transporter 1 transporter 1
Systematic nomenclature SLC7A7 SLC7A6 SLC7A9 SLC7A10 SLC7A11 SLC7A13
Common abbreviation y+LAT1 y+LAT2 b0,+ AT Asc-1 xCT –
HGNC, UniProt SLC7A7, Q9UM01 SLC7A6, Q92536 SLC7A9, P82251 SLC7A10, Q9NS82 SLC7A11, Q9UPY5 SLC7A13, Q8TCU3
Inhibitors – – – – quisqualate (pIC50 5.3) [188] –
Comments: CAT4 appears to be non-functional in heterologous Heterodimers between 4F2hc and y+ LAT1 or y+ LAT2 generate Asc-1 appears to heterodimerize with 4F2hc to allow the trans-
expression [672], while SLC7A14 has yet to be characterized. transporters similar to the system y+ L , which transport cationic port of small neutral amino acids (such as L-alanine, L-serine,
Glycoprotein-associated amino acid transporters are 12 TM pro- (L-arginine, L-lysine, L-ornithine) amino acids independent L-threonine, L-glutamine and glycine), as well as D-serine, in a
teins, which heterodimerize with members of the SLC3 family to of sodium and neutral (L-leucine, L-isoleucine, L-methionine, sodium-independent manner.
act as cell-surface amino acid exchangers. L-glutamine) amino acids in a partially sodium-dependent man- xCT generates a heterodimer with 4F2hc for a system x- e-c
Heterodimers between 4F2hc and LAT1 or LAT2 generate sodium- ner. These transporters are N-ethylmaleimide-insensitive. Het- transporter that mediates the sodium-independent exchange of
independent system L transporters. LAT1 transports large neutral erodimers between rBAT and b0,+ AT appear to mediate sodium- L-cystine and L-glutamic acid.
amino acids including branched-chain and aromatic amino acids independent system b0,+ transport of most of the neutral AGT has been conjugated with SLC3 members as fusion proteins
as well as miglustat, whereas LAT2 transports most of the neutral amino acids and cationic amino acids (L-arginine, L-lysine and to generate functional transporters, but the identity of a native
amino acids. L-ornithine). heterodimer has yet to be ascertained.
Further reading on SLC3 and SLC7 families of heteromeric amino acid transporters (HATs)
Bhutia YD et al. (2015) Amino Acid transporters in cancer and their relevance to "glutamine addic- Palacín M et al. (2005) The genetics of heteromeric amino acid transporters. Physiology (Bethesda) 20:
tion": novel targets for the design of a new class of anticancer drugs. Cancer Res. 75: 1782-8 112-24 [PMID:15772300]
[PMID:25855379] Verrey F et al. (2004) CATs and HATs: the SLC7 family of amino acid transporters. Pflugers Arch. 447:
Fotiadis D et al. (2013) The SLC3 and SLC7 families of amino acid transporters. Mol. Aspects Med. 532-42 [PMID:14770310]
34: 139-58 [PMID:23506863]
Palacín M et al. (2004) The ancillary proteins of HATs: SLC3 family of amino acid transporters.
Pflugers Arch. 447: 490-4 [PMID:14770309]
Overview: Together with the SLC26 family, the SLC4 family of Within the family, subgroups of transporters are identifiable: the gests a dimeric or tetrameric arrangement, with subunits made up
transporters subserve anion exchange, principally of chloride and electroneutral sodium-independent Cl- /HCO3 - transporters (AE1, of 13 TM domains and re-entrant loops at TM9/10 and TM11/12.
bicarbonate (HCO3 - ), but also carbonate and hydrogen sulphate AE2 and AE3), the electrogenic sodium-dependent HCO3 - trans- The N terminus exhibits sites for interaction with multiple pro-
(HSO4 - ). SLC4 family members regulate bicarbonate fluxes as part porters (NBCe1 and NBCe2) and the electroneutral HCO3 - trans- teins, including glycolytic enzymes, haemoglobin and cytoskele-
of carbon dioxide movement, chyme neutralization and reabsorp- porters (NBCn1 and NBCn2). Topographical information derives tal elements.
tion in the kidney. mainly from study of AE1, abundant in erythrocytes, which sug-
Anion exchangers
Transporters → SLC superfamily of solute carriers → SLC4 family of bicarbonate transporters → Anion exchangers
Nomenclature Anion exchange protein 1 Anion exchange protein 2 Anion exchange protein 3 Anion exchange protein 4
Systematic nomenclature SLC4A1 SLC4A2 SLC4A3 SLC4A9
Common abbreviation AE1 AE2 AE3 AE4
HGNC, UniProt SLC4A1, P02730 SLC4A2, P04920 SLC4A3, P48751 SLC4A9, Q96Q91
Endogenous substrates HCO3 -, Cl- Cl- , HCO3 - Cl- , HCO3 - –
Stoichiometry 1 Cl- (in) : 1 HCO3 - (out) 1 Cl- (in) : 1 HCO3 - (out) 1 Cl- (in) : 1 HCO3 - (out) –
Nomenclature Electrogenic sodium Electrogenic sodium Electroneutral sodium Electroneutral sodium NBCBE NaBC1
bicarbonate cotransporter 1 bicarbonate cotransporter 4 bicarbonate cotransporter 1 bicarbonate cotransporter 2
Systematic nomenclature SLC4A4 SLC4A5 SLC4A7 SLC4A10 SLC4A8 SLC4A11
Common abbreviation NBCe1 NBCe2 NBCn1 NBCn2 NDCBE BTR1
HGNC, UniProt SLC4A4, Q9Y6R1 SLC4A5, Q9BY07 SLC4A7, Q9Y6M7 SLC4A10, Q6U841 SLC4A8, Q2Y0W8 SLC4A11, Q8NBS3
Endogenous substrates NaHCO3 - NaHCO3 - NaHCO3 - NaHCO3 - NaHCO3 - , Cl- Cl- , NaHCO3 -
Stoichiometry 1 Na+ : 2/3 HCO3 - (out) or 1 1 Na+ : 2/3 HCO3 - (out) or 1 1 Na+ : 1 HCO3 - (out) or 1 1Na+ -
: 1 HCO3 (out) or 1 Na 1 Na+ -
: 2HCO3 (in) : 1 Cl- –
Na+ : CO3 2* Na+ : CO3 2* Na+ : CO3 2* : CO3 2* (out)
Majumdar D et al. (2010) Na-coupled bicarbonate transporters of the solute carrier 4 family in the Romero MF et al. (2013) The SLC4 family of bicarbonate (HCO_3− ) transporters. Mol. Aspects Med.
nervous system: function, localization, and relevance to neurologic function. Neuroscience 171: 34: 159-82 [PMID:23506864]
951-72 [PMID:20884330] Thornell IM et al. (2015) Regulators of Slc4 bicarbonate transporter activity. Front Physiol 6: 166
Parker MD et al. (2013) The divergence, actions, roles, and relatives of sodium-coupled bicarbonate [PMID:26124722]
transporters. Physiol. Rev. 93: 803-959 [PMID:23589833]
Reithmeier RA et al. (2016) Band 3, the human red cell chloride/bicarbonate anion exchanger (AE1,
SLC4A1), in a structural context. Biochim. Biophys. Acta 1858: 1507-32 [PMID:27058983]
Overview: Detailed characterisation of members of the hexose pressed in the small intestine, mediating the absorption of glucose reabsorption of glucose. SGLT3 is not a transporter but instead
transporter family is limited to SGLT1, 2 and 3, which are all in- (e.g. D-glucose), but also occurs in the brain, heart and in the late acts as a glucosensor generating an inwardly directed flux of Na+
hibited in a competitive manner by phlorizin, a natural dihydro- proximal straight tubule of the kidney. The expression of SGLT2 that causes membrane depolarization [153].
choline glucoside, that exhibits modest selectivity towards SGLT2 is almost exclusively restricted to the early proximal convoluted
(see [674] for an extensive review). SGLT1 is predominantly ex- tubule of the kidney, where it is largely responsible for the renal
Nomenclature Sodium/glucose cotransporter 1 Sodium/glucose cotransporter 2 Low affinity sodium-glucose Sodium/glucose cotransporter 4 Sodium/glucose cotransporter 5
cotransporter
Systematic nomenclature SLC5A1 SLC5A2 SLC5A4 SLC5A9 SLC5A10
Common abbreviation SGLT1 SGLT2 SGLT3 SGLT4 SGLT5
HGNC, UniProt SLC5A1, P13866 SLC5A2, P31639 SLC5A4, Q9NY91 SLC5A9, Q2M3M2 SLC5A10, A0PJK1
Substrates D-galactose [650], α-MDG [650], α-MDG, D-glucose D-glucose [650], D-glucose, D-mannose, α-MDG D-galactose, D-glucose
D-glucose [650] 1-deoxynojirimycin-1-sulfonic acid
[650], N-ethyl-1-deoxynojirimycin
[650], miglustat [650], miglitol
[650], 1-deoxynojirimycin [650]
Stoichiometry 2 Na+ : 1 glucose [340] 1 Na+ : 1 glucose [301] – – –
Selective inhibitors mizagliflozin (pKi 7.6) [311] dapagliflozin (pIC50 9.3) [343] – – –
Comments – – SGLT3 acts as a glucosensor. – –
Comments: Recognition and transport of substrate by SGLTs re- and C3, is tolerated (see [674] for a detailed quantification). Al- iological conditions [301]. Selective blockers of SGLT2, and thus
quires that the sugar is a pyranose. De-oxyglucose derivatives have though SGLT1 and SGLT2 have been described as high- and low- blocking 50% of renal glucose reabsorption, are in development
reduced affinity for SGLT1, but the replacement of the sugar equa- affinity sodium glucose co-transporters, respectively, recent work for the treatment of diabetes (e.g. [100]).
torial hydroxyl group by fluorine at some positions, excepting C2 suggests that they have a similar affinity for glucose under phys-
Choline transporter
Transporters → SLC superfamily of solute carriers → SLC5 family of sodium-dependent glucose transporters → Choline transporter
Overview: The high affinity, hemicholinium-3-sensitive, choline hydrolysis of ACh by acetylcholinesterase, CHT serves to maintain remains to be identified at the molecular level may involve multi-
transporter (CHT) is expressed mainly in cholinergic neurones on acetylcholine synthesis within the presynaptic terminal [200]. Ho- ple transporters. In addition, a family of choline transporter-like
nerve cell terminals and synaptic vesicles (keratinocytes being an mozygous mice engineered to lack CHT die within one hour of (CTL) proteins, (which are members of the SLC44 family) with
additional location). In autonomic neurones, expression of CHT birth as a result of hypoxia arising from failure of transmission at weak Na+ dependence have been described [622].
requires an activity-dependent retrograde signal from postsynap- the neuromuscular junction of the skeletal muscles that support
tic neurones [375]. Through recapture of choline generated by the respiration [199]. A low affinity choline uptake mechanism that
Nomenclature CHT
Systematic nomenclature SLC5A7
HGNC, UniProt SLC5A7, Q9GZV3
Substrates triethylcholine
Endogenous substrates choline
Stoichiometry Na+ : choline (variable stoichimetry); modulated by extracellular Cl- [322]
Selective inhibitors hemicholinium-3 (pKi 7–8) [478]
Labelled ligands [3 H]hemicholinium-3 (pKd 8.2–8.4)
Comments: Ki and KD values for hemicholinium-3 listed in the table are for human CHT expressed in Xenopus laevis oocytes [479], or COS-7 cells [22]. Hemicholinium mustard is a substrate for CHT
that causes covalent modification and irreversible inactivation of the transporter. Several exogenous substances (e.g. triethylcholine) that are substrates for CHT act as precursors to cholinergic false
transmitters.
Overview: The sodium-iodide symporter (NIS) is an iodide trans- (SLC5A8), which transports a wide range of monocarboxylates, is the kidney, SMCT2 is responsible for the bulk absorption of lactate
porter found principally in the thyroid gland where it mediates expressed in the apical membrane of epithelia of the small in- because of its low-affinity/high-capacity nature. Absence of both
the accumulation of I- within thyrocytes. Transport of I- by NIS testine, colon, kidney, brain neurones and the retinal pigment transporters in the kidney leads to massive excretion of lactate in
from the blood across the basolateral membrane followed by api- epithelium [224]. SMCT2 (SLC5A12) also localises to the apical urine and consequently drastic decrease in the circulating levels
cal efflux into the colloidal lumen, mediated at least in part by membrane of kidney, intestine, and colon, but in the brain and of lactate in blood [615]. SMCT1 also functions as a tumour sup-
pendrin (SLC22A4), and most likely not SMCT1 (SLC5A8) as once retina is restricted to astrocytes and Müller cells, respectively [224]. pressor in the colon as well as in various other non-colonic tissues
thought, provides the I- required for the synthesis of the thyroid SMCT1 is a high-affinity transporter whereas SMCT2 is a low- [225]. The tumour-suppressive function of SMCT1 is based on its
hormones triiodothyronine (triiodothyronine) and thyroxine (T4 ) affinity transporter. The physiological substrates for SMCT1 and ability to transport pyruvic acid, an inhibitor of histone deacety-
[59]. NIS is also expressed in the salivary glands, gastric mucosa, SMCT2 are lactate (L-lactic acid and D-lactic acid), pyruvic acid, lases, into cells in non-colonic tissues [616]; in the colon, the abil-
intestinal enterocytes and lactating breast. NIS mediates I- ab- propanoic acid, and nicotinic acid in non-colonic tissues such as ity of SMCT1 to transport butyric acid and propanoic acid, also in-
sorption in the intestine and I- secretion into the milk. SMVT is the kidney. SMCT1 is also likely to be the principal transporter for hibitors of histone deacetylases, underlies the tumour-suppressive
expressed on the apical membrane of intestinal enterocytes and the absorption of nicotinic acid (vitamin B3 ) in the intestine and function of this transporter [224, 225, 271]. The ability of SMCT1
colonocytes and is the main system responsible for biotin (vi- kidney [246]. In the small intestine and colon, the physiological to promote histone acetylase inhibition through accumulation of
tamin H) and pantothenic acid (vitamin B5 ) uptake in humans substrates for these transporters are nicotinic acid and the short- butyric acid and propanoic acid in immune cells is also respon-
[544]. SMVT located in kidney proximal tubule epithelial cells chain fatty acids acetic acid, propanoic acid, and butyric acid that sible for suppression of dendritic cell development in the colon
mediates the reabsorption of biotin and pantothenic acid. SMCT1 are produced by bacterial fermentation of dietary fiber [447]. In [579].
Comments: I- , ClO4 - , thiocyanate and NO3 - are competitive substrate inhibitors of NIS [157]. Lipoic acid appears to act as a competitive substrate inhibitor of SMVT [654] and the anticonvulsant drugs
primidone and carbamazepine competitively block the transport of biotin by brush border vesicles prepared from human intestine [545].
Overview: Three different mammalian myo-inositol cotrans- Darby canine kidney cell line, they segregate to the basolateral from the filtrate. In some species (e.g. rat, but not rabbit) apically
porters are currently known; two are the Na+ -coupled SMIT1 and and apical membranes, respectively [58]. In the nephron, SMIT1 located SMIT2 is responsible for the uptake of myo-inositol from
SMIT2 tabulated below and the third is proton-coupled HMIT mediates myo-inositol uptake as a ‘compatible osmolyte’ when in- the intestinal lumen [21].
(SLC2A13). SMIT1 and SMIT2 have a widespread and overlap- ner medullary tubules are exposed to increases in extracellular os-
ping tissue location but in polarized cells, such as the Madin- molality, whilst SMIT2 mediates the reabsorption of myo-inositol
Comments: The data tabulated are those for dog SMIT1 and rabbit SMIT2. SMIT2 transports D-chiro-inositol, but SMIT1 does not. In addition, whereas SMIT1 transports both D-xylose and L-xylose
and D-fucose and L-fucose, SMIT2 transports only the D-isomers of these sugars [121, 273]. Thus the substrate specificities of SMIT1 (for L-fucose) and SMIT2 (for D-chiro-inositol) allow discrimination
between the two SMITs. Human SMIT2 appears not to transport glucose [402].
DeFronzo RA et al. (2017) Renal, metabolic and cardiovascular considerations of SGLT2 inhibition. Wright EM. (2013) Glucose transport families SLC5 and SLC50. Mol. Aspects Med. 34: 183-96
Nat Rev Nephrol 13: 11-26 [PMID:27941935] [PMID:23506865]
Koepsell H. (2017) The Na+ -D-glucose cotransporters SGLT1 and SGLT2 are targets for the treatment Wright EM et al. (2011) Biology of human sodium glucose transporters. Physiol. Rev. 91: 733-94
of diabetes and cancer. Pharmacol. Ther. 170: 148-165 [PMID:27773781] [PMID:21527736]
Lehmann A et al. (2016) Intestinal SGLT1 in metabolic health and disease. Am. J. Physiol. Gastrointest.
Liver Physiol. 310: G887-98 [PMID:27012770]
Overview: Members of the solute carrier family 6 (SLC6) of monoamines, GABA, glycine and neutral amino acids, plus the LeuTAa , a Na+ -dependent amino acid transporter from Aquiflex ae-
sodium- and (sometimes chloride-) dependent neurotransmitter related bacterial NSS transporters [546]. The members of this su- olicus [687] and in several other transporter families structurally
transporters [80, 106, 376] are primarily plasma membrane lo- perfamily share a structural motif of 10 TM segments that has related to LeuT [209].
cated and may be divided into four subfamilies that transport been observed in crystal structures of the NSS bacterial homolog
Comments: [125 I]RTI55 labels all three monoamine transporters (NET, DAT and SERT) with affinities between 0.5 and 5 nM. Cocaine is an inhibitor of all three transporters with pKi values between 6.5
and 7.2. Potential alternative splicing sites in non-coding regions of SERT and NET have been identified. A bacterial homologue of SERT shows allosteric modulation by selected anti-depressants [580].
Overview: The activity of GABA-transporters located predomi- [566]. GAT1 is the predominant GABA transporter in the brain and is a high affinity taurine transporter involved in osmotic balance
nantly upon neurones (GAT-1), glia (GAT-3) or both (GAT-2, BGT- occurs primarily upon the terminals of presynaptic neurones and that occurs in the brain and non-neuronal tissues, such as the kid-
1) serves to terminate phasic GABA-ergic transmission, maintain to a much lesser extent upon distal astocytic processes that are in ney, brush border membrane of the intestine and blood brain bar-
low ambient extracellular concentrations of GABA, and recycle proximity to axons terminals. GAT3 resides predominantly on dis- rier [106, 279]. CT1, which transports creatine, has a ubiquitous
GABA for reuse by neurones. Nonetheless, ambient concentra- tal astrocytic terminals that are close to the GABAergic synapse. By expression pattern, often co-localizing with creatine kinase [106].
tions of GABA are sufficient to sustain tonic inhibition mediated contrast, BGT1 occupies an extrasynaptic location possibly along
by high affinity GABAA receptors in certain neuronal populations with GAT2 which has limited expression in the brain [422]. TauT
Comments: The IC50 values for GAT1-4 reported in the table re- [218]. In addition to the inhibitors listed, deramciclane is a take of GABA [see [119, 561] for reviews]. GAT3 is inhibited by
flect the range reported in the literature from studies of both hu- moderately potent, though non-selective, inhibitor of all cloned physiologically relevant concentrations of Zn2+ [123]. Taut trans-
man and mouse transporters. There is a tendency towards lower GABA transporters (IC50 = 26-46 μM; [148]). Diaryloxime and ports GABA, but with low affinity, but CT1 does not, although
IC50 values for the human orthologue [380]. SNAP-5114 is only diarylvinyl ether derivatives of nipecotic acid and guvacine that it can be engineered to do so by mutagenesis guided by LeuT
weakly selective for GAT 2 and GAT3, with IC50 values in the
potently inhibit the uptake of [3 H]GABA into rat synaptosomes as a structural template [155]. Although inhibitors of creatine
range 22 to >30 μM at GAT1 and BGT1, whereas NNC052090
has at least an order of magnitude selectivity for BGT1 [see have been described [359]. Several derivatives of exo-THPO (e.g. transport by CT1 (e.g. β-guanidinopropionic acid, cyclocreatine,
[119, 562] for reviews]. Compound (R)-4d is a recently described N-methyl-exo-THPO and N-acetyloxyethyl-exo-THPO) demon- guanidinoethane sulfonic acid) are known (e.g. [131]) they insuf-
compound that displays 20-fold selectivity for GAT3 over GAT1 strate selectivity as blockers of astroglial, versus neuronal, up- ficiently characterized to be included in the table.
Overview: Two gene products, GlyT1 and GlyT2, are known that gous mice engineered to totally lack GlyT1 exhibit severe respi- second postnatal week [244] and mutations in the human gene
give rise to transporters that are predominantly located on glia and ratory and motor deficiencies due to hyperactive glycinergic sig- encoding GlyT2 (SLC6A5) have been identified in patients with
neurones, respectively. Five variants of GlyT1 (a,b,c,d & e) differ- nalling and die within the first postnatal day [243, 624]. Dis- hyperekplexia (reviewed by [281]). ATB0+ (SLC6A14) is a trans-
ing in their N- and C-termini are generated by alternative pro- ruption of GlyT1 restricted to forebrain neurones is associated porter for numerous dipolar and cationic amino acids and thus
moter usage and splicing, and three splice variants of GlyT2 (a,b with enhancement of EPSCs mediated by NMDA receptors and
has a much broader substrate specificity than the glycine trans-
& c) have also been identified (see [51, 189, 242, 594] for reviews). behaviours that are suggestive of a promnesic action [695]. GlyT2
porters alongside which it is grouped on the basis of structural
GlyT1 transporter isoforms expressed in glia surrounding gluta- transporters localised on the axons and boutons of glycinergic
matergic synapses regulate synaptic glycine concentrations in- neurones appear crucial for efficient transmitter loading of synap- similarity [106]. ATB0+ is expressed in various peripheral tissues
fluencing NMDA receptor-mediated neurotransmission [50, 219], tic vesicles but may not be essential for the termination of in- [106]. By contrast PROT (SLC6A7), which is expressed only in
but also are important, in early neonatal life, for regulating glycine hibitory neurotransmission [244, 537]. Mice in which GlyT2 has brain in association with a subset of excitatory nerve terminals,
concentrations at inhibitory glycinergic synapses [243]. Homozy- been deleted develop a fatal hyperekplexia phenotype during the shows specificity for the transport of L-proline.
Comments: Sarcosine is a selective transportable inhibitor of vary, most likely due to differences in assay conditions [74, 424]. and N-arachidonyl-D-alanine have been described as endogenous
GlyT1 and also a weak agonist at the glycine binding site of the The tricyclic antidepressant amoxapine weakly inhibits GlyT2 non-competitive inhibitors of GlyT2a, but not GlyT1b [170, 327,
NMDA receptor [707], but has no effect on GlyT2. This differ- (IC50 92 μM) with approximately 10-fold selectivity over GlyT1 668]. Protons [30] and Zn2+ [332] act as non-competitive in-
ence has been attributed to a single glycine residue in TM6 (ser- [473]. The endogenous lipids arachidonic acid and anandamide hibitors of GlyT1b, with IC50 values of 100 nM and 10 μM respec-
ine residue in GlyT2) [641]. Inhibition of GLYT1 by the sarco- exert opposing effects upon GlyT1a, inhibiting (IC50 2 μM) tively, but neither ion affects GlyT2 (reviewed by [639]). Glycine
sine derivatives NFPS, NPTS and Org 24598 is non-competitive and potentiating (EC50 13 μM) transport currents, respectively transport by GLYT1 is inhibited by Li+ , whereas GLYT2 transport
[424, 436]. IC50 values for Org 24598 reported in the literature [491]. N-arachidonyl-glycine, N-arachidonyl-γ-aminobutyric acid is stimulated (both in the presence of Na+ ) [509].
Bermingham DP et al. (2016) Kinase-dependent Regulation of Monoamine Neurotransmitter Trans- Joncquel-Chevalier Curt M et al. (2015) Creatine biosynthesis and transport in health and disease.
porters. Pharmacol. Rev. 68: 888-953 [PMID:27591044] Biochimie 119: 146-65 [PMID:26542286]
Bröer S et al. (2012) The solute carrier 6 family of transporters. Br. J. Pharmacol. 167: 256-78 Lohr KM et al. (2017) Membrane transporters as mediators of synaptic dopamine dynamics: impli-
[PMID:22519513] cations for disease. Eur. J. Neurosci. 45: 20-33 [PMID:27520881]
Overview: The sodium/calcium exchangers (NCX) use the extra- ATPase (SERCA), as well as the sodium/potassium/calcium ex- tion to allow calcium influx and sodium efflux, as an electrogenic
changers (NKCX, SLC24 family), NCX allow recovery of intracel- mechanism. Structural modelling suggests the presence of 9 TM
cellular sodium concentration to facilitate the extrusion of cal-
lular calcium back to basal levels after cellular stimulation. When segments, with a large intracellular loop between the fifth and
cium out of the cell. Alongside the plasma membrane Ca2+ - intracellular sodium ion levels rise, for example, following de- sixth TM segments.
ATPase (PMCA) and sarcoplasmic/endoplasmic reticulum Ca2+ - polarisation, these transporters can operate in the reverse direc-
Comments: Although subtype-selective inhibitors of NCX function are not widely available, 3,4-dichlorobenzamil and CBDMB act as non-selective NCX inhibitors, while SEA0400, KB-R7943, SN6, and
ORM-10103 [331] act to inhibit NCX function with varying degrees of selectivity. BED is a selective NCX3 inhibitor [563] and and YM-244769 inhibits NCX3 preferentially over other isoforms [323, 688].
Giladi M et al. (2016) Structure-Functional Basis of Ion Transport in Sodium-Calcium Exchanger Sekler I. (2015) Standing of giants shoulders the story of the mitochondrial Na(+)Ca(2+) exchanger.
(NCX) Proteins. Int J Mol Sci 17: [PMID:27879668] Biochem. Biophys. Res. Commun. 460: 50-2 [PMID:25998733]
Khananshvili D. (2013) The SLC8 gene family of sodium-calcium exchangers (NCX) - structure, func-
tion, and regulation in health and disease. Mol. Aspects Med. 34: 220-35 [PMID:23506867]
Overview: Sodium/hydrogen exchangers or sodium/proton an- NHE9 appear to locate on intracellular membranes [448, 457, 472]. NHE1 is considered to be a ubiquitously-expressed ‘housekeeping’
tiports are a family of transporters that maintain cellular pH by Li+ and NH4 + , but not K+ , ions may also be transported by some transporter. NHE3 is highly expressed in the intestine and kid-
utilising the sodium gradient across the plasma membrane to ex- isoforms. Modelling of the topology of these transporters indi- neys and regulate sodium movements in those tissues. NHE10 is
trude protons produced by metabolism, in a stoichiometry of 1 cates 12 TM regions with an extended intracellular C-terminus present in sperm [653] and osteoclasts [391]; gene disruption re-
Na+ (in) : 1 H+ (out). Several isoforms, NHE6, NHE7, NHE8 and containing multiple regulatory sites. sults in infertile male mice [653].
Comments: Analogues of the non-selective cation transport inhibitor amiloride appear to inhibit NHE function through competitive inhibition of the extracellular Na+ binding site. The more selective
amiloride analogues MPA and ethylisopropylamiloride exhibit a rank order of affinity of inhibition of NHE1 > NHE2 > NHE3 [127, 625, 626].
Donowitz M et al. (2013) SLC9/NHE gene family, a plasma membrane and organellar family of Parker MD et al. (2015) Na+-H+ exchanger-1 (NHE1) regulation in kidney proximal tubule. Cell. Mol.
Na+ /H+ exchangers. Mol. Aspects Med. 34: 236-51 [PMID:23506868] Life Sci. 72: 2061-74 [PMID:25680790]
Kato A et al. (2011) Regulation of electroneutral NaCl absorption by the small intestine. Annu. Rev. Ruffin VA et al. (2014) Intracellular pH regulation by acid-base transporters in mammalian neurons.
Physiol. 73: 261-81 [PMID:21054167] Front Physiol 5: 43 [PMID:24592239]
Ohgaki R et al. (2011) Organellar Na+/H+ exchangers: novel players in organelle pH regulation and
their emerging functions. Biochemistry 50: 443-50 [PMID:21171650]
Overview: The SLC10 family transport bile acids, sulphated maintain the enterohepatic circulation of bile acids [138, 357]. tity of their endogenous substrates remain unknown [201, 236,
solutes, and other xenobiotics in a sodium-dependent manner. SLC10A6 (SOAT) functions as a sodium-dependent transporter 241, 649]. Members of the SLC10 family are predicted to have
The founding members, SLC10A1 (NTCP) and SLC10A2 (ASBT) of sulphated solutes including sulfphated steroids and bile acids seven transmembrane domains with an extracellular N-terminus
function, along with members of the ABC transporter family [234, 236]. Transport function has not yet been demonstrated and cytoplasmic C-terminus [42, 274].
(MDR1/ABCB1, BSEP/ABCB11 and MRP2/ABCC2) and the organic for the 4 remaining members of the SLC10 family, SLC10A3 (P3),
solute transporter obligate heterodimer OSTα:OSTβ (SLC51), to SLC10A4 (P4), SLC10A5 (P5), and SLC10A7 (P7), and the iden-
Nomenclature Sodium/bile acid and sulphated solute cotransporter 1 Sodium/bile acid and sulphated solute cotransporter 2 Sodium/bile acid and sulphated solute cotransporter 6
Systematic nomenclature SLC10A1 SLC10A2 SLC10A6
Common abbreviation NTCP ASBT SOAT
HGNC, UniProt SLC10A1, Q14973 SLC10A2, Q12908 SLC10A6, Q3KNW5
Substrates – glycodeoxycholic acid > glycoursodeoxycholic acid, pregnenolone sulphate [234], estrone-3-sulphate,
glycochenodeoxycholic acid > taurocholic acid > dehydroepiandrosterone sulphate [236],
cholic acid [129] taurolithocholic acid-3-sulphate
Endogenous substrates dehydroepiandrosterone sulphate [129, 201, 430], – –
estrone-3-sulphate, iodothyronine sulphates [649]
tauroursodeoxycholic acid, taurocholic acid,
taurochenodeoxycholic acid > glycocholic acid >
cholic acid [430]
Stoichiometry 2 Na+ : 1 bile acid [42, 234] >1 Na+ : 1 bile acid [129, 662] –
Inhibitors (-)-propranolol (pIC50 8.2) [355], cyclosporin A (pIC50 6) elobixibat (pIC50 8.9) [237], SC-435 (pIC50 8.8) [54], –
[355], (+)-propranolol (pIC50 5.3) [355], cyclosporin A (pKi 264W94 (pIC50 7.3) [620, 679]
5.1) [159], irbesartan (pKi 4.9) [159]
Labelled ligands – [3 H]taurocholic acid [129] –
Comments – Chenodeoxycholyl-N -nitrobenzoxadiazol-lysine is a –
fluorescent bile acid analogue used as a probe [662].
Comments: Heterologously expressed SLC10A4 [235] or SLC10A7 [241] failed to exhibit significant transport of taurocholic acid, pregnenolone sulphate, dehydroepiandrosterone sulphate or choline.
SLC10A4 has recently been suggested to associate with neuronal vesicles [84].
Anwer MS et al. (2014) Sodium-dependent bile salt transporters of the SLC10A transporter family: Dawson PA. (2017) Roles of Ileal ASBT and OSTα-OSTβ in Regulating Bile Acid Signaling. Dig Dis 35:
more than solute transporters. Pflugers Arch. 466: 77-89 [PMID:24196564] 261-266 [PMID:28249269]
Claro da Silva T et al. (2013) The solute carrier family 10 (SLC10): beyond bile acid transport. Mol. Zwicker BL et al. (2013) Transport and biological activities of bile acids. Int. J. Biochem. Cell Biol. 45:
Aspects Med. 34: 252-69 [PMID:23506869] 1389-98 [PMID:23603607]
Overview: The family of proton-coupled metal ion trans- dent on proton transport. Both proteins appear to have 12 TM re- the pathogen. Alternatively, export of divalent cations from the
porters are responsible for movements of divalent cations, par- gions and cytoplasmic N- and C- termini. NRAMP1 is involved in phagosome may deprive the pathogen of essential enzyme cofac-
ticularly ferrous and manganese ions, across the cell membrane antimicrobial action in macrophages, although its precise mech- tors. SLC11A2/DMT1 is more widely expressed and appears to
(SLC11A2/DMT1) and across endosomal (SLC11A2/DMT1) or anism is undefined. Facilitated diffusion of divalent cations into assist in divalent cation assimilation from the diet, as well as in
lysosomal/phagosomal membranes (SLC11A1/NRAMP1), depen- phagosomes may increase intravesicular free radicals to damage phagocytotic cells.
Comments: Loss-of-function mutations in NRAMP1 are associated with increased susceptibility to microbial infection (OMIM: 607948). Loss-of-function mutations in DMT1 are associated with microcytic
anemia (OMIM: 206100).
Codazzi F et al. (2015) Iron entry in neurons and astrocytes: a link with synaptic activity. Front Mol Wessling-Resnick M. (2015) Nramp1 and Other Transporters Involved in Metal Withholding during
Neurosci 8: 18 [PMID:26089776] Infection. J. Biol. Chem. 290: 18984-90 [PMID:26055722]
Montalbetti N et al. (2013) Mammalian iron transporters: families SLC11 and SLC40. Mol. Aspects Zheng W et al. (2012) Regulation of brain iron and copper homeostasis by brain barrier systems:
Med. 34: 270-87 [PMID:23506870] implication in neurodegenerative diseases. Pharmacol. Ther. 133: 177-88 [PMID:22115751]
Overview: The SLC12 family of chloride transporters contribute thiazide diuretics. These 12 TM proteins exhibit cytoplasmic ter- tended extracellular loop at TM 5/6. CCC6 exhibits structural sim-
to ion fluxes across a variety of tissues, particularly in the kidney mini and an extended extracellular loop at TM7/8 and are kidney- ilarities with the K-Cl co-transporters, while CCC9 is divergent,
and choroid plexus of the brain. Within this family, further sub- specific (NKCC2 and NCC) or show a more widespread distribu- with 11 TM domains and a cytoplasmic N-terminus and extracel-
families are identifiable: NKCC1, NKCC2 and NCC constitute a tion (NKCC1). A second family, the K-Cl co-transporters are also lular C-terminus.
group of therapeutically-relevant transporters, targets for loop and 12 TM domain proteins with cytoplasmic termini, but with an ex-
Nomenclature Kidney-specific Na-K-Cl symporter Basolateral Na-K-Cl symporter Na-Cl symporter K-Cl cotransporter 1 K-Cl cotransporter 2
Systematic nomenclature SLC12A1 SLC12A2 SLC12A3 SLC12A4 SLC12A5
Common abbreviation NKCC2 NKCC1 NCC KCC1 KCC2
HGNC, UniProt SLC12A1, Q13621 SLC12A2, P55011 SLC12A3, P55017 SLC12A4, Q9UP95 SLC12A5, Q9H2X9
Stoichiometry 1 Na+ : 1 K+ : 2 Cl- (in) 1 Na+ : 1 K+ : 2 Cl- (in) 1 Na+ : 1 Cl- (in) 1 K+ : 1 Cl- (out) 1 K+ : 1 Cl- (out)
Inhibitors bumetanide (pIC50 6.5) [280], piretanide (pIC50 5.6) [280], chlorothiazide, cyclothiazide, DIOA VU0240551 (pIC50 6.2) [143],
piretanide (pIC50 6) [280], bumetanide (pIC50 5.6) [280], hydrochlorothiazide, metolazone DIOA
furosemide (pIC50 5.2) [280] furosemide (pIC50 5.1) [280]
Comments: DIOA is able to differentiate KCC isoforms from NKCC and NCC transporters, but also inhibits CFTR [321].
Arroyo JP et al. (2013) The SLC12 family of electroneutral cation-coupled chloride cotransporters. Huang X et al. (2016) Everything we always wanted to know about furosemide but were afraid to
Mol. Aspects Med. 34: 288-98 [PMID:23506871] ask. Am. J. Physiol. Renal Physiol. 310: F958-71 [PMID:26911852]
Bachmann S et al. (2017) Regulation of renal Na-(K)-Cl cotransporters by vasopressin. Pflugers Arch. Kahle KT et al. (2015) K-Cl cotransporters, cell volume homeostasis, and neurological disease. Trends
469: 889-897 [PMID:28577072] Mol Med 21: 513-23 [PMID:26142773]
Bazúa-Valenti S et al. (2016) Physiological role of SLC12 family members in the kidney. Am. J. Physiol. Martín-Aragón Baudel MA et al. (2017) Chloride co-transporters as possible therapeutic targets for
Renal Physiol. 311: F131-44 [PMID:27097893] stroke. J. Neurochem. 140: 195-209 [PMID:27861901]
Nomenclature Na+ /sulfate cotransporter Na+ /dicarboxylate cotransporter 1 Na+ /dicarboxylate cotransporter 3 Na+ /sulfate cotransporter Na+ /citrate cotransporter
Systematic nomenclature SLC13A1 SLC13A2 SLC13A3 SLC13A4 SLC13A5
Common abbreviation NaS1 NaC1 NaC3 NaS2 NaC2
HGNC, UniProt SLC13A1, Q9BZW2 SLC13A2, Q13183 SLC13A3, Q8WWT9 SLC13A4, Q9UKG4 SLC13A5, Q86YT5
Endogenous substrates SeO4 2- , SO4 2- , S2 O3 2- citric acid, succinic acid citric acid, succinic acid SO4 2- citric acid, pyruvic acid
Stoichiometry 3 Na+ : 1 SO4 2- (in) 3 Na+ : 1 dicarboxylate2- (in) Unknown 3 Na+ : SO4 2- (in) Unknown
Bergeron MJ et al. (2013) SLC13 family of Na+ -coupled di- and tri-carboxylate/sulfate transporters. Pajor AM. (2014) Sodium-coupled dicarboxylate and citrate transporters from the SLC13 family.
Mol. Aspects Med. 34: 299-312 [PMID:23506872] Pflugers Arch. 466: 119-30 [PMID:24114175]
Markovich D. (2014) Na+-sulfate cotransporter SLC13A1. Pflugers Arch. 466: 131-7 [PMID:24193406]
Overview: As a product of protein catabolism, urea is moved its concentration gradient. Multiple splice variants of these trans- mains, with a glycosylated extracellular loop at TM5/6, and intra-
around the body and through the kidneys for excretion. Although porters have been identified; for UT-A transporters, in particular, cellular C- and N-termini. The UT-A1 splice variant, exceptionally,
there is experimental evidence for concentrative urea transporters, there is evidence for cell-specific expression of these variants with has 20 TM domains, equivalent to a combination of the UT-A2 and
these have not been defined at the molecular level. The SLC14 functional impact [589]. Topographical modelling suggests that UT-A3 splice variants.
family are facilitative transporters, allowing urea movement down the majority of the variants of SLC14 transporters have 10 TM do-
Esteva-Font C et al. (2015) Urea transporter proteins as targets for small-molecule diuretics. Nat Rev Shayakul C et al. (2013) The urea transporter family (SLC14): physiological, pathological and struc-
Nephrol 11: 113-23 [PMID:25488859] tural aspects. Mol. Aspects Med. 34: 313-22 [PMID:23506873]
LeMoine CM et al. (2015) Evolution of urea transporters in vertebrates: adaptation to urea’s multiple Stewart G. (2011) The emerging physiological roles of the SLC14A family of urea transporters. Br. J.
roles and metabolic sources. J. Exp. Biol. 218: 1936-1945 [PMID:26085670] Pharmacol. 164: 1780-92 [PMID:21449978]
Pannabecker TL. (2013) Comparative physiology and architecture associated with the mammalian
urine concentrating mechanism: role of inner medullary water and urea transport pathways in
the rodent medulla. Am. J. Physiol. Regul. Integr. Comp. Physiol. 304: R488-503 [PMID:23364530]
Overview: The Solute Carrier 15 (SLC15) family of peptide digestion, SLC15A2 (PEPT2) mainly allows renal tubular reup- transporters variably interacts with a very large number of pep-
transporters, alias H+ -coupled oligopeptide cotransporter family, take of di/tripeptides from ultrafiltration and brain-to-blood ef- tidomimetics and peptide-like drugs. It is conceivable, based on
is a group of membrane transporters known for their key role flux of di/tripeptides in the choroid plexus, SLC15A3 (PHT2) and the currently acknowledged structural and functional differences,
in the cellular uptake of di- and tripeptides (di/tripeptides). Of SLC15A4 (PHT1) interact with both di/tripeptides and histidine, to divide the SLC15 family of peptide transporters into two sub-
its members, SLC15A1 (PEPT1) chiefly mediates intestinal ab- e.g. in certain immune cells, and SLC15A5 has unknown phys- families.
sorption of luminal di/tripeptides from overall dietary protein iological function. In addition, the SLC15 family of peptide
Comments: The members of the SLC15 family of peptide with the SLC15 family transporters. Known substrates include of Gly-Sar by SLC15A1 and/or SLC15A2 [483].
transporters are particularly promiscuous in the transport of cefadroxil, valacyclovir, 5-aminolevulinic acid, L-Dopa prodrugs, There is evidence to suggest the existence of a fifth member of
di/tripeptides, and D-amino acid containing peptides are also gemcitabine prodrugs, floxuridine prodrugs, Maillard reaction this transporter family, SLC15A5 (A6NIM6; ENSG00000188991),
transported. While SLC15A3 and SLC15A4 transport histi- products, JBP485, zanamivir, oseltamivir prodrugs, doxorubicin but to date there is no established biological function or reported
dine, none of them transport tetrapeptides. In addition, many prodrugs, polymyxins, and didanosine prodrugs. Frequently used pharmacology for this protein [585].
molecules, among which beta-lactam antibiotics, angiotensin- pharmaceutical excipients such as Tween® 20, Tween® 80, Solu-
converting enzyme inhibitors and sartans, variably interact tol® HS 15 and Cremophor EL® strongly inhibit cellular uptake
Anderson CM et al. (2010) Hijacking solute carriers for proton-coupled drug transport. Physiology Newstead S. (2017) Recent advances in understanding proton coupled peptide transport via the POT
(Bethesda) 25: 364-77 [PMID:21186281] family. Curr. Opin. Struct. Biol. 45: 17-24 [PMID:27865112]
Brandsch M. (2013) Drug transport via the intestinal peptide transporter PepT1. Curr Opin Pharmacol Newstead S. (2011) Towards a structural understanding of drug and peptide transport within the
13: 881-7 [PMID:24007794] proton-dependent oligopeptide transporter (POT) family. Biochem. Soc. Trans. 39: 1353-8
Brandsch M. (2009) Transport of drugs by proton-coupled peptide transporters: pearls and pitfalls. [PMID:21936814]
Expert Opin Drug Metab Toxicol 5: 887-905 [PMID:19519280]
Fredriksson R et al. (2008) The solute carrier (SLC) complement of the human genome: phylogenetic Smith DE et al. (2013) Proton-coupled oligopeptide transporter family SLC15: physiological, phar-
classification reveals four major families. FEBS Lett. 582: 3811-6 [PMID:18948099] macological and pathological implications. Mol. Aspects Med. 34: 323-36 [PMID:23506874]
Newstead S. (2015) Molecular insights into proton coupled peptide transport in the PTR family of Thwaites DT et al. (2007) H+-coupled nutrient, micronutrient and drug transporters in the mam-
oligopeptide transporters. Biochim. Biophys. Acta 1850: 488-499 [PMID:24859687] malian small intestine. Exp. Physiol. 92: 603-19 [PMID:17468205]
Comments: MCT1 and MCT2, but not MCT3 and MCT4, are inhibited by CHC, which also inhibits members of the mitochondrial transporter family, SLC25.
MCT5-MCT7, MCT9 and MCT11-14 are regarded as orphan transporters.
Bernal J et al. (2015) Thyroid hormone transporters-functions and clinical implications. Nat Rev Jones RS et al. (2016) Monocarboxylate Transporters: Therapeutic Targets and Prognostic Factors in
Endocrinol 11: 406-417 [PMID:25942657] Disease. Clin. Pharmacol. Ther. 100: 454-463 [PMID:27351344]
Halestrap AP. (2013) The SLC16 gene family - structure, role and regulation in health and disease.
Mol. Aspects Med. 34: 337-49 [PMID:23506875]
Nomenclature Sodium/phosphate cotransporter 1 Sodium/phosphate cotransporter 3 Sodium/phosphate cotransporter 4 Sodium/phosphate cotransporter homolog
Systematic nomenclature SLC17A1 SLC17A2 SLC17A3 SLC17A4
Common abbreviation NPT1 NPT3 NPT4 –
HGNC, UniProt SLC17A1, Q14916 SLC17A2, O00624 SLC17A3, O00476 SLC17A4, Q9Y2C5
Substrates probenecid [86], penicillin G [86], Cl- [306], – – –
organic acids [306], uric acid [306],
phosphate [306]
Stoichiometry Unknown Unknown Unknown Unknown
Nomenclature Sialin
Systematic nomenclature SLC17A5
Common abbreviation AST
HGNC, UniProt SLC17A5, Q9NRA2
Endogenous substrates L-lactic acid, gluconate (out), L-glutamic acid (in) [446], glucuronic acid, L-aspartic acid [446], sialic acid
Stoichiometry 1 H+ : 1 sialic acid (out)
Comments: Loss-of-function mutations in sialin are associated with Salla disease (OMIM: 604369), an autosomal recessive neurodegenerative disorder associated with sialic acid storage disease [647].
Nomenclature Vesicular glutamate transporter 1 Vesicular glutamate transporter 2 Vesicular glutamate transporter 3
Systematic nomenclature SLC17A7 SLC17A6 SLC17A8
Common abbreviation VGLUT1 VGLUT2 VGLUT3
HGNC, UniProt SLC17A7, Q9P2U7 SLC17A6, Q9P2U8 SLC17A8, Q8NDX2
Endogenous substrates L-glutamic acid > D-glutamic acid L-glutamic acid > D-glutamic acid L-glutamic acid > D-glutamic acid
Stoichiometry Unknown Unknown Unknown
Comments: Endogenous ketoacids produced during fasting have been proposed to regulate VGLUT function through blocking chloride ion-mediated allosteric enhancement of transporter function
[333].
Comments: VGLUTs and VNUT can be inhibited by DIDS and evans blue dye.
Moriyama Y et al. (2017) Vesicular nucleotide transporter (VNUT): appearance of an actress on the Reimer RJ. (2013) SLC17: a functionally diverse family of organic anion transporters. Mol. Aspects
stage of purinergic signaling. Purinergic Signal. 13: 387-404 [PMID:28616712] Med. 34: 350-9 [PMID:23506876]
Omote H et al. (2016) Structure, Function, and Drug Interactions of Neurotransmitter Transporters Takamori S. (2016) Vesicular glutamate transporters as anion channels? Pflugers Arch. 468: 513-8
in the Postgenomic Era. Annu. Rev. Pharmacol. Toxicol. 56: 385-402 [PMID:26514205] [PMID:26577586]
Overview: The vesicular amine transporters (VATs) are puta- multi-subunit vacuolar ATPase that acidifies secretory vesicles (re- [183] distributes between both central and peripheral sympathetic
tive 12 TM domain proteins that function to transport singly viewed by [174]). The vesicular acetylcholine transporter (VAChT; monoaminergic neurones [175]. The vescular polyamine trans-
positively charged amine neurotransmitters and hormones from [184]) localizes to cholinergic neurons, but non-neuronal expres- porter (VPAT) is highly expressed in the lungs and placenta, with
the cytoplasm and concentrate them within secretory vesicles. sion has also been claimed [558]. Vesicular monoamine trans- moderate expression in brain and testis, and with low expression
They function as amine/proton antiporters driven by secondary porter 1 (VMAT1, [182]) is mainly expressed in peripheral neu- in heart and skeletal muscle [290]. VPAT mediates vesicular accu-
active transport utilizing the proton gradient established by a roendocrine cells, but most likely not in the CNS, whereas VMAT2 mulation of polyamines in mast cells [602].
Nomenclature Vesicular monoamine transporter 1 Vesicular monoamine transporter 2 Vesicular acetylcholine transporter
Systematic nomenclature SLC18A1 SLC18A2 SLC18A3
Common abbreviation VMAT1 VMAT2 VAChT
HGNC, UniProt SLC18A1, P54219 SLC18A2, Q05940 SLC18A3, Q16572
Substrates dexamfetamine (Ki 4.7×10−5 M) [183], β-phenylethylamine β-phenylethylamine (Ki 3.7×10−6 M) [183], dexamfetamine TPP+ [71], ethidium [71], N-methyl-pyridinium-2-aldoxime
(Ki 3.4×10−5 M) [183], fenfluramine (Ki 3.1×10−6 M) [183], (Ki 2.1×10−6 M) [183], fenfluramine (Ki 5.1×10−6 M) [183], [71],
MPP+ (Ki 6.9×10−5 M) [183], MDMA (Ki 1.9×10−5 M) [183] MPP+ (Ki 8.9×10−6 M) [183], MDMA (Ki 6.9×10−6 M) [183] N-(4’-pentanonyl)-4-(4”-dimethylamino-styryl)pyridinium
[71]
Endogenous substrates histamine (Ki 4.6×10−3 M) [183], 5-hydroxytryptamine (Ki histamine (Ki 1.4×10−4 M) [183], dopamine (Ki acetylcholine (Ki 7.9×10−4 M) [72, 352], choline (Ki
1.4×10−6 M) [183], dopamine (Ki 3.8×10−6 M) [183], 1.4×10−6 M) [183], 5-hydroxytryptamine (Ki 9×10−7 M) 5×10−3 M) [72, 352]
(-)-noradrenaline (Ki 1.3×10−5 M) [183], (-)-adrenaline (Ki [183], (-)-noradrenaline (Ki 3.4×10−6 M) [183],
5.5×10−6 M) [183] (-)-adrenaline (Ki 1.9×10−6 M) [183]
Stoichiometry 1 amine (in): 2H+ (out) 1 amine (in): 2H+ (out) 1 amine (in): 2H+ (out)
Inhibitors reserpine (pKi 7.5) [183], ketanserin (pKi 5.8) [183], reserpine (pKi 7.9) [183], tetrabenazine (pKi 7) [183], aminobenzovesamicol (pKi 10.9) [173], vesamicol (pKi 8.7)
tetrabenazine (pKi 4.7) [183] ketanserin (pKi 6.3) [183] [173]
Labelled ligands – [3 H]TBZOH (Inhibitor) (pKd 8.2) [644], [125 I]iodovinyl-TBZ [3 H]vesamicol (pKd 8.4) [644], [123 I]iodobenzovesamicol
(Inhibitor) (pKd 8.1) [378], [11 C]DTBZ (Inhibitor),
[125 I]7-azido-8-iodoketanserine (Inhibitor) [577]
Comments: pKi values for endogenous and synthetic substrate inhibitors of human VMAT1 and VMAT2 are for inhibition of [3 H]5-HT uptake in transfected and permeabilised CV-1 cells as detailed
by [183]. In addition to the monoamines listed in the table, the trace amines tyramine and β-phenylethylamine are probable substrates for VMAT2 [175]. Probes listed in the table are those currently
employed; additional agents have been synthesized (e.g. [720]).
German CL et al. (2015) Regulation of the Dopamine and Vesicular Monoamine Transporters: Phar- Sitte HH et al. (2015) Amphetamines, new psychoactive drugs and the monoamine transporter cycle.
macological Targets and Implications for Disease. Pharmacol. Rev. 67: 1005-24 [PMID:26408528] Trends Pharmacol. Sci. 36: 41-50 [PMID:25542076]
Lohr KM et al. (2017) Membrane transporters as mediators of synaptic dopamine dynamics: impli- Wimalasena K. (2011) Vesicular monoamine transporters: structure-function, pharmacology, and
cations for disease. Eur. J. Neurosci. 45: 20-33 [PMID:27520881] medicinal chemistry. Med Res Rev 31: 483-519 [PMID:20135628]
Omote H et al. (2016) Structure, Function, and Drug Interactions of Neurotransmitter Transporters
in the Postgenomic Era. Annu. Rev. Pharmacol. Toxicol. 56: 385-402 [PMID:26514205]
Comments: Loss-of-function mutations in ThTr1 underlie thiamine-responsive megaloblastic anemia syndrome [151].
Matherly LH et al. (2014) The major facilitative folate transporters solute carrier 19A1 and solute Zhao R et al. (2013) Folate and thiamine transporters mediated by facilitative carriers (SLC19A1-3
carrier 46A1: biology and role in antifolate chemotherapy of cancer. Drug Metab. Dispos. 42: and SLC46A1) and folate receptors. Mol. Aspects Med. 34: 373-85 [PMID:23506878]
632-49 [PMID:24396145]
Biber J et al. (2013) Phosphate transporters and their function. Annu. Rev. Physiol. 75: 535-50 Shobeiri N et al. (2014) Phosphate: an old bone molecule but new cardiovascular risk factor. Br J Clin
[PMID:23398154] Pharmacol 77: 39-54 [PMID:23506202]
Forster IC et al. (2013) Phosphate transporters of the SLC20 and SLC34 families. Mol. Aspects Med.
34: 386-95 [PMID:23506879]
Searchable database: http://www.guidetopharmacology.org/index.jsp SLC22 family of organic cation and anion transporters S443
Full Contents of ConciseGuide: http://onlinelibrary.wiley.com/doi/10.1111/bph.14753/full
S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: Transporters. British Journal of Pharmacology (2019) 176, S397–S493
Nomenclature Organic cation transporter 1 Organic cation transporter 2 Organic cation transporter 3
Systematic nomenclature SLC22A1 SLC22A2 SLC22A3
Common abbreviation OCT1 OCT2 OCT3
HGNC, UniProt SLC22A1, O15245 SLC22A2, O15244 SLC22A3, O75751
Substrates MPP+ , tetraethylammonium, desipramine, metformin MPP+ [247], pancuronium [247], tetraethylammonium MPP+ , tetraethylammonium, quinidine, metformin [368]
[576], aciclovir [247], tubocurarine [247], cisplatin [368], metformin [368]
Endogenous substrates PGF2α , choline, PGE2 , 5-hydroxytryptamine PGE2 [356], dopamine [263], histamine [263] (-)-noradrenaline [719], 5-hydroxytryptamine [719],
dopamine [719]
Stoichiometry Unknown Unknown Unknown
Inhibitors clonidine (pKi 6.3) [708] decynium 22 (pKi 7) [247] disprocynium24 (pKi 7.8) [264]
Comments: Corticosterone and quinine are able to inhibit all three organic cation transporters.
Koepsell H. (2013) The SLC22 family with transporters of organic cations, anions and zwitterions. Pelis RM et al. (2014) SLC22, SLC44, and SLC47 transporters–organic anion and cation transporters:
Mol. Aspects Med. 34: 413-35 [PMID:23506881] molecular and cellular properties. Curr Top Membr 73: 233-61 [PMID:24745985]
Lozano E et al. (2013) Role of the plasma membrane transporter of organic cations OCT1 and its Yin J et al. (2016) Renal drug transporters and their significance in drug-drug interactions. Acta
genetic variants in modern liver pharmacology. Biomed Res Int 2013: 692071 [PMID:23984399] Pharm Sin B 6: 363-373 [PMID:27709005]
Comments: Mutations in the SLC22A5 gene lead to primary carnitine deficiency [409].
Pochini L et al. (2013) OCTN cation transporters in health and disease: role as drug targets and assay Yin J et al. (2016) Renal drug transporters and their significance in drug-drug interactions. Acta
development. J Biomol Screen 18: 851-67 [PMID:23771822] Pharm Sin B 6: 363-373 [PMID:27709005]
Tamai I. (2013) Pharmacological and pathophysiological roles of carnitine/organic cation trans-
porters (OCTNs: SLC22A4, SLC22A5 and Slc22a21). Biopharm Drug Dispos 34: 29-44
[PMID:22952014]
Nomenclature Organic anion transporter 1 Organic anion transporter 2 Organic anion transporter 3 Organic anion transporter 4 Organic anion transporter 5 Organic anion transporter 7
Systematic nomenclature SLC22A6 SLC22A7 SLC22A8 SLC22A11 SLC22A10 SLC22A9
Common abbreviation OAT1 OAT2 OAT3 – OAT5 OAT4
HGNC, UniProt SLC22A6, Q4U2R8 SLC22A7, Q9Y694 SLC22A8, Q8TCC7 SLC22A11, Q9NSA0 SLC22A10, Q63ZE4 SLC22A9, Q8IVM8
Substrates aminohippuric acid, aminohippuric acid, PGE2 , uric acid [466], uric acid [466], ochratoxin A [697] –
non-steroidal non-steroidal estrone-3-sulphate [379], dehydroepiandrosterone sulphate
anti-inflammatory drugs anti-inflammatory drugs aminohippuric acid [379], [97], estrone-3-sulphate
cimetidine [379], [97], ochratoxin A [97]
ochratoxin A [379]
Stoichiometry Unknown Unknown Unknown Unknown Unknown Unknown
Inhibitors probenecid (Inhibition of – – – – –
urate transport by human
SCL22A6.) (pIC50 4.9) [304]
Burckhardt G et al. (2011) In vitro and in vivo evidence of the importance of organic anion trans- Shen H et al. (2017) Organic Anion Transporter 2: An Enigmatic Human Solute Carrier. Drug Metab.
porters (OATs) in drug therapy. Handb Exp Pharmacol 29-104 [PMID:21103968] Dispos. 45: 228-236 [PMID:27872146]
Koepsell H. (2013) The SLC22 family with transporters of organic cations, anions and zwitterions. Yin J et al. (2016) Renal drug transporters and their significance in drug-drug interactions. Acta
Mol. Aspects Med. 34: 413-35 [PMID:23506881] Pharm Sin B 6: 363-373 [PMID:27709005]
Urate transporter
Transporters → SLC superfamily of solute carriers → SLC22 family of organic cation and anion transporters → Urate transporter
Overview: URAT1, a member of the OAT (organic anion transporter) family, is an anion-exchanging uptake transporter localized to the apical (brush border) membrane of renal proximal tubular cells.
It is an anion exchanger that specifically reabsorbs uric acid from the proximal tubule in exchange for monovalent anions such as lactate, nicotinoate, acetoacetate, and hydroxybutyrate [181].
Nigam SK et al. (2018) The systems biology of uric acid transporters: the role of remote sensing and
signaling. Curr. Opin. Nephrol. Hypertens. 27: 305-313 [PMID:29847376]
Comments: There are three human synaptic vesicle glycoprotein 2 family members, SV2A, SV2B and SV2C. They have transmembrane transporter activity and can be classified in to the SLC superfamily
of solute carriers in subfamily SLC22, as SCL22B1, B2 and B3 respectively. SV2A (SCL22B1) has been identified as the brain binding-site for the antiepileptic drugs levetiracetam [358, 415] and brivaracetam
[465].
Löscher W et al. (2016) Synaptic Vesicle Glycoprotein 2A Ligands in the Treatment of Epilepsy and Mendoza-Torreblanca JG et al. (2013) Synaptic vesicle protein 2A: basic facts and role in synaptic
Beyond. CNS Drugs 30: 1055-1077 [PMID:27752944] function. Eur. J. Neurosci. 38: 3529-39 [PMID:24102679]
Burckhardt G. (2012) Drug transport by Organic Anion Transporters (OATs). Pharmacol. Ther. 136: Lozano E et al. (2018) Genetic Heterogeneity of SLC22 Family of Transporters in Drug Disposition. J
106-30 [PMID:22841915] Pers Med 8: [PMID:29659532]
Hillgren KM et al. (2013) Emerging transporters of clinical importance: an update from the Interna- Nigam SK. (2018) The SLC22 Transporter Family: A Paradigm for the Impact of Drug Transporters
tional Transporter Consortium. Clin. Pharmacol. Ther. 94: 52-63 [PMID:23588305] on Metabolic Pathways, Signaling, and Disease. Annu. Rev. Pharmacol. Toxicol. 58: 663-687
International Transporter Consortium et al. (2010) Membrane transporters in drug development. [PMID:29309257]
Nat Rev Drug Discov 9: 215-36 [PMID:20190787] Zamek-Gliszczynski MJ et al. (2018) Transporters in Drug Development: 2018 ITC Recommenda-
Koepsell H. (2013) The SLC22 family with transporters of organic cations, anions and zwitterions. tions for Transporters of Emerging Clinical Importance. Clin. Pharmacol. Ther. 104: 890-899
Mol. Aspects Med. 34: 413-35 [PMID:23506881] [PMID:30091177]
Nomenclature Sodium-dependent vitamin C transporter 1 Sodium-dependent vitamin C transporter 2 Sodium-dependent vitamin C transporter 3
Systematic nomenclature SLC23A1 SLC23A2 SLC23A3
Common abbreviation SVCT1 SVCT2 SVCT3
HGNC, UniProt SLC23A1, Q9UHI7 SLC23A2, Q9UGH3 SLC23A3, Q6PIS1
Endogenous substrates L-ascorbic acid > D-ascorbic acid > dehydroascorbic acid L-ascorbic acid > D-ascorbic acid > dehydroascorbic acid –
[628] [628]
Stoichiometry 2 Na+ : 1 ascorbic acid (in) [628] 2 Na+ : 1 ascorbic acid (in) [628] –
Inhibitors phloretin (pKi 4.2) [628] – –
Labelled ligands [14 C]ascorbic acid (Binding) [417] [14 C]ascorbic acid –
Comments – – SLC23A3 does not transport ascorbic acid and remains an
orphan transporter.
Bürzle M et al. (2013) The sodium-dependent ascorbic acid transporter family SLC23. Mol. Aspects May JM. (2011) The SLC23 family of ascorbate transporters: ensuring that you get and keep your
Med. 34: 436-54 [PMID:23506882] daily dose of vitamin C. Br. J. Pharmacol. 164: 1793-801 [PMID:21418192]
Comments: NKCX6 has been proposed to be the sole member of a CAX Na+ /Ca2+ exchanger family, which may be the mitochondrial transporter responsible for calcium accumulation from the cytosol
[565].
Schnetkamp PP. (2013) The SLC24 gene family of Na+ /Ca2+ -K+ exchangers: from sight and smell to Sekler I. (2015) Standing of giants shoulders the story of the mitochondrial Na(+)Ca(2+) exchanger.
memory consolidation and skin pigmentation. Mol. Aspects Med. 34: 455-64 [PMID:23506883] Biochem. Biophys. Res. Commun. 460: 50-2 [PMID:25998733]
Schnetkamp PP et al. (2014) The SLC24 family of K+ -dependent Na+ -Ca2+ exchangers: structure-
function relationships. Curr Top Membr 73: 263-87 [PMID:24745986]
Nomenclature Mitochondrial citrate transporter Mitochondrial dicarboxylate transporter Mitochondrial oxoglutarate carrier Mitochondrial oxodicarboxylate carrier
Systematic nomenclature SLC25A1 SLC25A10 SLC25A11 SLC25A21
Common abbreviation CIC DIC OGC ODC
HGNC, UniProt SLC25A1, P53007 SLC25A10, Q9UBX3 SLC25A11, Q02978 SLC25A21, Q9BQT8
Substrates phosphoenolpyruvic acid, malic acid, 2- ,
SO4 phosphate, S2 O3 2- , succinic acid, α-ketoglutaric acid, malic acid α-ketoglutaric acid, α-oxoadipic acid
citric acid malic acid
Stoichiometry Malate2- (in) : H-citrate2- (out) PO3 4- (in) : malate2- (out) Malate2- (in) : oxoglutarate2- (out) Oxoadipate (in) : oxoglutarate (out)
Inhibitors 1,2,3-benzenetricarboxylic acid – – –
Searchable database: http://www.guidetopharmacology.org/index.jsp Mitochondrial di- and tri-carboxylic acid transporter subfamily S450
Full Contents of ConciseGuide: http://onlinelibrary.wiley.com/doi/10.1111/bph.14753/full
S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: Transporters. British Journal of Pharmacology (2019) 176, S397–S493
Nomenclature AGC1 AGC2 Mitochondrial Mitochondrial Mitochondrial ornithine Mitochondrial ornithine Carnitine/acylcarnitine
glutamate carrier 2 glutamate carrier 1 transporter 2 transporter 1 carrier
Systematic nomenclature SLC25A12 SLC25A13 SLC25A18 SLC25A22 SLC25A2 SLC25A15 SLC25A20
Common abbreviation – – GC2 GC1 ORC2 ORC1 CAC
HGNC, UniProt SLC25A12, O75746 SLC25A13, Q9UJS0 SLC25A18, Q9H1K4 SLC25A22, Q9H936 SLC25A2, Q9BXI2 SLC25A15, Q9Y619 SLC25A20, O43772
Substrates L-glutamic acid, 2-amino-3- L-glutamic acid L-glutamic acid L-citrulline [202], L-lysine [202], –
2-amino-3- sulfinopropanoic acid, L-arginine [202], L-ornithine [202],
sulfinopropanoic acid, L-glutamic acid, L-lysine [202], D-lysine L-citrulline [202],
L-aspartic acid L-aspartic acid [202], D-arginine [202], L-arginine [202]
D-citrulline [202],
D-ornithine [202],
L-ornithine [202],
D-histidine [202],
L-histidine [202]
Stoichiometry Aspartate : glutamate Aspartate : glutamate Glutamate : H+ Glutamate : H+ 1 Ornithine (in) :1 1 Ornithine (in) :1 –
H+ (bidirectional) H+ (bidirectional) (bidirectional) (bidirectional) citrulline : 1 H+ (out) citrulline : 1 H+ (out)
Comments – – – – – – Exchanges cytosolic
acylcarnitine for
mitochondrial carnitine
Comments: Both ornithine transporters are inhibited by the polyamine spermine [203]. Loss-of-function mutations in these genes are associated with hyperornithinemia-hyperammonemia-homocitrullinuria.
Nomenclature Mitochondrial Mitochondrial Mitochondrial Mitochondrial Graves disease carrier Peroxisomal membrane
adenine nucleotide adenine nucleotide adenine nucleotide adenine nucleotide protein
translocator 1 translocator 2 translocator 3 translocator 4
Systematic nomenclature SLC25A4 SLC25A5 SLC25A6 SLC25A31 SLC25A16 SLC25A17
Common abbreviation ANT1 ANT2 ANT3 ANT4 GDC PMP34
HGNC, UniProt SLC25A4, P12235 SLC25A5, P05141 SLC25A6, P12236 SLC25A31, Q9H0C2 SLC25A16, P16260 SLC25A17, O43808
Substrates – – – – CoA and congeners ADP, ATP,
adenosine 5’-monophosphate
Stoichiometry ADP3- (in) : ATP4- (out) ADP3- (in) : ATP4- (out) ADP3- (in) : ATP4- (out) ADP3- (in) : ATP4- (out) CoA (in) ATP (in)
Inhibitors bongkrek acid, – – – – –
carboxyatractyloside
Nomenclature Deoxynucleotide carrier 1 S-Adenosylmethionine carrier Mitochondrial phosphate carrier 1 Mitochondrial phosphate carrier 2 Mitochondrial phosphate carrier 3
Systematic nomenclature SLC25A19 SLC25A26 SLC25A24 SLC25A23 SLC25A25
Common abbreviation DNC SAMC1 APC1 APC2 APC3
HGNC, UniProt SLC25A19, Q9HC21 SLC25A26, Q70HW3 SLC25A24, Q6NUK1 SLC25A23, Q9BV35 SLC25A25, Q6KCM7
Substrates Nucleotide Diphosphates (NDPs), S-adenosyl methionine – – –
Deoxynucleotide Diphosphates
(dNDPs), Dideoxynucleotide
Triphosphates (ddNTPs),
Deoxynucleotide Triphosphates
(dNTPs)
Stoichiometry dNDP (in) : ATP (out) – – – –
Nomenclature Uncoupling protein 1 Uncoupling protein 2 Uncoupling protein 3 Uncoupling protein 4 Uncoupling protein 5 KMCP1
Systematic nomenclature SLC25A7 SLC25A8 SLC25A9 SLC25A27 SLC25A14 SLC25A30
Common abbreviation UCP1 UCP2 UCP3 UCP4 UCP5 –
HGNC, UniProt UCP1, P25874 UCP2, P55851 UCP3, P55916 SLC25A27, O95847 SLC25A14, O95258 SLC25A30, Q5SVS4
Stoichiometry H+ (in) H+ (in) H+ (in) H+ (in) H+ (in) –
Baffy G. (2017) Mitochondrial uncoupling in cancer cells: Liabilities and opportunities. Biochim. Palmieri F. (2013) The mitochondrial transporter family SLC25: identification, properties and phys-
Biophys. Acta 1858: 655-664 [PMID:28088333] iopathology. Mol. Aspects Med. 34: 465-84 [PMID:23266187]
Seifert EL et al. (2015) The mitochondrial phosphate carrier: Role in oxidative metabolism, cal-
Bertholet AM et al. (2017) UCP1: A transporter for H+ and fatty acid anions. Biochimie 134: 28-34
cium handling and mitochondrial disease. Biochem. Biophys. Res. Commun. 464: 369-75
[PMID:27984203]
[PMID:26091567]
Clémençon B et al. (2013) The mitochondrial ADP/ATP carrier (SLC25 family): pathological impli- Taylor EB. (2017) Functional Properties of the Mitochondrial Carrier System. Trends Cell Biol. 27:
cations of its dysfunction. Mol. Aspects Med. 34: 485-93 [PMID:23506884] 633-644 [PMID:28522206]
Chloride/bicarbonate exchangers
Transporters → SLC superfamily of solute carriers → SLC26 family of anion exchangers → Chloride/bicarbonate exchangers
Anion channels
Transporters → SLC superfamily of solute carriers → SLC26 family of anion exchangers → Anion channels
Nomenclature Prestin
Systematic nomenclature SLC26A5
HGNC, UniProt SLC26A5, P58743
Substrates HCO3 - [443], Cl- [443]
Stoichiometry Unknown
Comments Prestin has been suggested to function as a molecular motor, rather than a transporter
Alper SL et al. (2013) The SLC26 gene family of anion transporters and channels. Mol. Aspects Med. Nofziger C et al. (2011) Pendrin function in airway epithelia. Cell. Physiol. Biochem. 28: 571-8
34: 494-515 [PMID:23506885] [PMID:22116372]
Kato A et al. (2011) Regulation of electroneutral NaCl absorption by the small intestine. Annu. Rev. Soleimani M. (2013) SLC26 Cl-/HCO3- exchangers in the kidney: roles in health and disease. Kidney
Physiol. 73: 261-81 [PMID:21054167] Int. 84: 657-66 [PMID:23636174]
Overview: Fatty acid transporter proteins (FATPs) are a family brane associated domain, peripheral membrane associated do- chain acyl-CoA synthetase (EC 6.2.1.- , EC 6.2.1.7) enzyme activ-
(SLC27) of six transporters (FATP1-6). They have at least one, main, FATP signature, intracellular AMP binding motif, dimer- ity. Within the cell, these transporters may associate with plasma
and possibly six [397, 557], transmembrane segments, and are ization domain, lipocalin motif, and an ER localization domain and peroxisomal membranes. FATP1-4 and -6 transport long- and
predicted on the basis of structural similarities to form dimers. (identified in FATP4 only) [190, 441, 481]. These transporters very long-chain fatty acids, while FATP5 transports long-chain
SLC27 members have several structural domains: integral mem- are unusual in that they appear to express intrinsic very long- fatty acids as well as bile acids [439, 557].
Nomenclature Fatty acid transport protein 1 Fatty acid transport Fatty acid transport Fatty acid transport protein 4 Fatty acid transport Fatty acid transport
protein 2 protein 3 protein 5 protein 6
Systematic nomenclature SLC27A1 SLC27A2 SLC27A3 SLC27A4 SLC27A5 SLC27A6
Common abbreviation FATP1 FATP2 FATP3 FATP4 FATP5 FATP6
HGNC, UniProt SLC27A1, Q6PCB7 SLC27A2, O14975 SLC27A3, Q5K4L6 SLC27A4, Q6P1M0 SLC27A5, Q9Y2P5 SLC27A6, Q9Y2P4
Endogenous substrates palmitic acid > oleic acid > – – palmitic acid , oleic acid > γ-linolenic acid – palmitic acid >
γ-linolenic acid > octanoic acid [238] > octanoic acid [238] oleic acid >
arachidonic acid > palmitic acid > palmitic acid > oleic acid > butyric acid, γ-linolenic acid >
oleic acid > butyric acid [557] γ-linolenic acid > arachidonic acid [586] octanoic acid [238]
Inhibitors – – – compound 1l (pIC50 7.1) [60] – –
Comments – – – FATP4 is genetically linked to – –
restrictive dermopathy.
Comments: Although the stoichiometry of fatty acid transport [718], have been described; analysis of the mechanism of action of FATP2 has two variants: Variant 1 encodes the full-length protein,
is unclear, it has been proposed to be facilitated by the coupling some of these inhibitors suggests that transport may be selectively while Variant 2 encodes a shorter isoform missing an internal pro-
of fatty acid transport to conjugation with coenzyme A to form inhibited without altering enzymatic activity of the FATP. tein segment. FATP6 also has two variants: Variant 2 encodes the
fatty acyl CoA esters. Small molecule inhibitors of FATP2 [398, C1-BODIPY-C12 accumulation has been used as a non-selective same protein as Variant 1 but has an additional segment in the 5’
553] and FATP4 [60, 718], as well as bile acid inhibitors of FATP5 index of fatty acid transporter activity. UTR.
Anderson CM et al. (2013) SLC27 fatty acid transport proteins. Mol. Aspects Med. 34: 516-28 Schwenk RW et al. (2010) Fatty acid transport across the cell membrane: regulation by fatty acid
[PMID:23506886] transporters. Prostaglandins Leukot. Essent. Fatty Acids 82: 149-54 [PMID:20206486]
Dourlen P et al. (2015) Fatty acid transport proteins in disease: New insights from invertebrate mod-
els. Prog. Lipid Res. 60: 30-40 [PMID:26416577]
SLC28 family
Transporters → SLC superfamily of solute carriers → SLC28 and SLC29 families of nucleoside transporters → SLC28 family
Overview: SLC28 family membersappear to have 13 TM segments with cytoplasmic N-termini and extracellular C-termini, and function as concentrative nucleoside transporters.
SLC29 family
Transporters → SLC superfamily of solute carriers → SLC28 and SLC29 families of nucleoside transporters → SLC29 family
Overview: SLC29 family members appear to be composed of 11 TM segments with cytoplasmic N-termini and extracellular C-termini. ENT1, ENT2 and ENT4 are cell-surface transporters, while ENT3 is
intracellular, possibly lysosomal [38]. ENT1-3 are described as broad-spectrum equilibrative nucleoside transporters, while ENT4 is primarily a polyspecific organic cation transporter at neutral pH [293].
Boswell-Casteel RC et al. (2017) Equilibrative nucleoside transporters-A review. Nucleosides Nu- Wang J. (2016) The plasma membrane monoamine transporter (PMAT): Structure, function, and role
cleotides Nucleic Acids 36: 7-30 [PMID:27759477] in organic cation disposition. Clin. Pharmacol. Ther. 100: 489-499 [PMID:27506881]
Pastor-Anglada M et al. (2018) Who Is Who in Adenosine Transport. Front Pharmacol 9: 627
[PMID:29962948]
Boswell-Casteel RC et al. (2017) Equilibrative nucleoside transporters-A review. Nucleosides Nu- Young JD. (2016) The SLC28 (CNT) and SLC29 (ENT) nucleoside transporter families: a 30-year col-
cleotides Nucleic Acids 36: 7-30 [PMID:27759477] laborative odyssey. Biochem. Soc. Trans. 44: 869-76 [PMID:27284054]
Pastor-Anglada M et al. (2015) Nucleoside transporter proteins as biomarkers of drug responsiveness Young JD et al. (2013) The human concentrative and equilibrative nucleoside transporter families,
and drug targets. Front Pharmacol 6: 13 [PMID:25713533] SLC28 and SLC29. Mol. Aspects Med. 34: 529-47 [PMID:23506887]
Overview: Along with the SLC39 family, SLC30 transporters reg- membrane extruding zinc, while ZnT3 is associated with synap- lent linking has been suggested as a mechanism for dimerisation,
ulate the movement of zinc ions around the cell. In particular, tic vesicles and ZnT4 and ZnT5 are linked with secretory granules. particularly for ZnT3 [551]. The mechanism for zinc transport is
these transporters remove zinc ions from the cytosol, allowing ac- Membrane topology predictions suggest a multimeric assembly, unknown.
cumulation into intracellular compartments or efflux through the potentially heteromultimeric [596], with subunits having six TM
plasma membrane. ZnT1 is thought to be placed on the plasma domains, and both termini being cytoplasmic. Dityrosine cova-
Bouron A et al. (2014) Contribution of calcium-conducting channels to the transport of zinc ions. Kambe T et al. (2014) Current understanding of ZIP and ZnT zinc transporters in human health and
Pflugers Arch. 466: 381-7 [PMID:23719866] diseases. Cell. Mol. Life Sci. 71: 3281-95 [PMID:24710731]
Hojyo S et al. (2016) Zinc transporters and signaling in physiology and pathogenesis. Arch. Biochem. Kambe T et al. (2015) The Physiological, Biochemical, and Molecular Roles of Zinc Transporters in
Biophys. 611: 43-50 [PMID:27394923] Zinc Homeostasis and Metabolism. Physiol. Rev. 95: 749-784 [PMID:26084690]
Huang L et al. (2013) The SLC30 family of zinc transporters - a review of current understanding of
their biological and pathophysiological roles. Mol. Aspects Med. 34: 548-60 [PMID:23506888]
Comments: Copper accumulation through CTR1 is sensitive to silver ions, but not divalent cations [387].
Howell SB et al. (2010) Copper transporters and the cellular pharmacology of the platinum- Kim H et al. (2013) SLC31 (CTR) family of copper transporters in health and disease. Mol. Aspects
containing cancer drugs. Mol. Pharmacol. 77: 887-94 [PMID:20159940] Med. 34: 561-70 [PMID:23506889]
Kaplan JH et al. (2016) How Mammalian Cells Acquire Copper: An Essential but Potentially Toxic Monné M et al. (2014) Antiporters of the mitochondrial carrier family. Curr Top Membr 73: 289-320
Metal. Biophys. J. 110: 7-13 [PMID:26745404] [PMID:24745987]
Overview: The vesicular inhibitory amino acid transporter, termini [429]. However, an alternative 9TM structure with the N that VIAAT is instead a Cl- /GABA co-transporter. VIAAT co-exists
VIAAT (also termed the vesicular GABA transporter VGAT), terminus facing the cytoplasm and the C terminus residing in the with VGLUT1 (SLC17A7), or VGLUT2 (SLC17A6), in the synaptic
which is the sole representative of the SLC32 family, trans- synaptic vesicle lumen has subsequently been reported [427]. VI- vesicles of selected nerve terminals [194, 701]. VIAAT knock out
ports GABA, or glycine, into synaptic vesicles [229, 230], and AAT acts as an antiporter for inhibitory amino acids and protons. mice die between embryonic day 18.5 and birth [671]. In cultures
is a member of the structurally-defined amino acid-polyamine- The accumulation of GABA and glycine within vesicles is driven by of spinal cord neurones established from earlier embryos, the co-
organocation/APC clan composed of SLC32, SLC36 and SLC38 both the chemical (pH) and electrical (ψ) components of the release of of GABA and glycine from synaptic vesicles is drastically
transporter families (see [559]). VIAAT was originally suggested proton electrochemical gradient (μH +) established by a vacuolar reduced, providing direct evidence for the role of VIAAT in the se-
to be composed of 10 TM segments with cytoplasmic N- and C- H+ -ATPase [429]. However, one study, [334], presented evidence questration of both transmitters [547, 671].
Searchable database: http://www.guidetopharmacology.org/index.jsp SLC32 vesicular inhibitory amino acid transporter S462
Full Contents of ConciseGuide: http://onlinelibrary.wiley.com/doi/10.1111/bph.14753/full
S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: Transporters. British Journal of Pharmacology (2019) 176, S397–S493
Anne C et al. (2014) Vesicular neurotransmitter transporters: mechanistic aspects. Curr Top Membr Schiöth HB et al. (2013) Evolutionary origin of amino acid transporter families SLC32, SLC36 and
73: 149-74 [PMID:24745982] SLC38 and physiological, pathological and therapeutic aspects. Mol. Aspects Med. 34: 571-85
[PMID:23506890]
Comments: In heterologous expression studies, acetyl CoA transport through AT1 was inhibited by coenzyme A, but not acetic acid, ATP or UDP-galactose [330]. A loss-of-function mutation in
ACATN1/SLC33A1 has been associated with spastic paraplegia (SPG42, [401]), although this observation could not be replicated in a subsequent study [560].
Hirabayashi Y et al. (2004) The acetyl-CoA transporter family SLC33. Pflugers Arch. 447: 760-2 Hirabayashi Y et al. (2013) The acetyl-CoA transporter family SLC33. Mol. Aspects Med. 34: 586-9
[PMID:12739170] [PMID:23506891]
Comments: These transporters can be inhibited by foscarnet, in contrast to type III sodium-phosphate cotransporters, the SLC20 family.
Biber J et al. (2013) Phosphate transporters and their function. Annu. Rev. Physiol. 75: 535-50 Shobeiri N et al. (2014) Phosphate: an old bone molecule but new cardiovascular risk factor. Br J Clin
[PMID:23398154] Pharmacol 77: 39-54 [PMID:23506202]
Forster IC et al. (2013) Phosphate transporters of the SLC20 and SLC34 families. Mol. Aspects Med. Wagner CA et al. (2014) The SLC34 family of sodium-dependent phosphate transporters. Pflugers
34: 386-95 [PMID:23506879] Arch. 466: 139-53 [PMID:24352629]
Nomenclature CMP-sialic acid transporter UDP-galactose transporter UDP-N-acetylglucosamine PAPS transporter 1 PAPS transporter 2
transporter
Systematic nomenclature SLC35A1 SLC35A2 SLC35A3 SLC35B2 SLC35B3
HGNC, UniProt SLC35A1, P78382 SLC35A2, P78381 SLC35A3, Q9Y2D2 SLC35B2, Q8TB61 SLC35B3, Q9H1N7
Substrates CMP-sialic acid [313] UDP-galactose [315, 445], UDP N-acetyl-glucosamine [316] A3P5PS [336] A3P5PS [335]
UDP N-acetyl-glucosamine [315,
445]
Ishida N et al. (2004) Molecular physiology and pathology of the nucleotide sugar transporter family Song Z. (2013) Roles of the nucleotide sugar transporters (SLC35 family) in health and disease. Mol.
(SLC35). Pflugers Arch. 447: 768-75 [PMID:12759756] Aspects Med. 34: 590-600 [PMID:23506892]
Orellana A et al. (2016) Overview of Nucleotide Sugar Transporter Gene Family Functions Across
Multiple Species. J. Mol. Biol. 428: 3150-3165 [PMID:27261257]
Overview: Members of the SLC36 family of proton-coupled expressed at the luminal surface of the small intestine and absorbs brown/beige adipocytes [633]. PAT1 and PAT4 are involved in reg-
amino acid transporters are involved in membrane transport of amino acids and derivatives [3]. In lysosomes, PAT1 functions as ulation of the mTORC1 pathway [191]. More comprehensive lists
amino acids and derivatives. The four transporters show variable an efflux mechanism for amino acids produced during intralysoso- of substrates can be found within the reviews under Further Read-
tissue expression patterns and are expressed in various cell types mal proteolysis [5, 542]. PAT2 is expressed at the apical membrane ing and in the references.
at the plasma-membrane and in intracellular organelles. PAT1 is of the renal proximal tubule [82] and at the plasma-membrane in
Searchable database: http://www.guidetopharmacology.org/index.jsp SLC36 family of proton-coupled amino acid transporters S466
Full Contents of ConciseGuide: http://onlinelibrary.wiley.com/doi/10.1111/bph.14753/full
S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: Transporters. British Journal of Pharmacology (2019) 176, S397–S493
Schiöth HB et al. (2013) Evolutionary origin of amino acid transporter families SLC32, SLC36 and Thwaites DT et al. (2007) Deciphering the mechanisms of intestinal imino (and amino)
SLC38 and physiological, pathological and therapeutic aspects. Mol. Aspects Med. 34: 571-85 acid transport: the redemption of SLC36A1. Biochim. Biophys. Acta 1768: 179-97
[PMID:23506890] [PMID:17123464]
Thwaites DT et al. (2011) The SLC36 family of proton-coupled amino acid transporters and their
potential role in drug transport. Br. J. Pharmacol. 164: 1802-16 [PMID:21501141]
Searchable database: http://www.guidetopharmacology.org/index.jsp SLC36 family of proton-coupled amino acid transporters S467
Full Contents of ConciseGuide: http://onlinelibrary.wiley.com/doi/10.1111/bph.14753/full
S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: Transporters. British Journal of Pharmacology (2019) 176, S397–S493
Chou JY et al. (2014) The SLC37 family of sugar-phosphate/phosphate exchangers. Curr Top Membr Chou JY et al. (2013) The SLC37 family of phosphate-linked sugar phosphate antiporters. Mol. As-
73: 357-82 [PMID:24745989] pects Med. 34: 601-11 [PMID:23506893]
Searchable database: http://www.guidetopharmacology.org/index.jsp SLC38 family of sodium-dependent neutral amino acid transporters S468
Full Contents of ConciseGuide: http://onlinelibrary.wiley.com/doi/10.1111/bph.14753/full
S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: Transporters. British Journal of Pharmacology (2019) 176, S397–S493
Nomenclature sodium-coupled neutral amino acid transporter 1 sodium-coupled neutral amino acid transporter 2 sodium-coupled neutral amino acid transporter 4
Systematic nomenclature SLC38A1 SLC38A2 SLC38A4
Common abbreviation SNAT1 SNAT2 SNAT4
HGNC, UniProt SLC38A1, Q9H2H9 SLC38A2, Q96QD8 SLC38A4, Q969I6
Substrates MeAIB MeAIB MeAIB
L-alanine > L-serine, L-glutamine, L-asparagine, L-histidine, L-alanine, L-methionine > L-asparagine, L-glutamine, L-histidine > L-arginine, L-alanine, L-asparagine, L-lysine >
L-cysteine, L-methionine > glycine, L-threonine, L-proline, L-serine, L-proline, glycine > L-threonine, L-leucine, glycine, L-glutamine, L-serine, L-proline, L-leucine,
L-tyrosine, L-valine [7] L-phenylalanine [283] L-phenylalanine [282]
Stoichiometry 1 Na+ : 1 amino acid (in) [7] 1 Na+ : 1 amino acid (in) [283] 1 Na+ : 1 neutral amino acid (in) [282]
Labelled ligands [14 C]alanine, [3 H]alanine [14 C]alanine, [3 H]alanine [14 C]alanine, [14 C]glycine, [3 H]alanine, [3 H]glycine
Comments – – Transport of cationic amino acids by SNAT4 was
sodium-independent [282].
Nomenclature Sodium-coupled neutral amino acid transporter 3 Sodium-coupled neutral amino acid transporter 5
Systematic nomenclature SLC38A3 SLC38A5
Common abbreviation SNAT3 SNAT5
HGNC, UniProt SLC38A3, Q99624 SLC38A5, Q8WUX1
Substrates MeAIB MeAIB
L-histidine , L-glutamine > L-asparagine, L-alanine > L-glutamic acid [197] L-asparagine, L-serine, L-histidine, L-glutamine > glycine, L-alanine [459]
Stoichiometry 1 Na+ : 1 amino acid (in) : 1 H+ (out) [75] 1 Na+ : 1 amino acid (in) : 1 H+ (out) [459]
Labelled ligands [14 C]glutamine, [3 H]glutamine [14 C]histidine, [3 H]histidine
Bhutia YD et al. (2016) Glutamine transporters in mammalian cells and their functions in physiology Hägglund MG et al. (2011) Identification of SLC38A7 (SNAT7) protein as a glutamine transporter
and cancer. Biochim. Biophys. Acta 1863: 2531-9 [PMID:26724577] expressed in neurons. J. Biol. Chem. 286: 20500-11 [PMID:21511949]
Bröer S. (2014) The SLC38 family of sodium-amino acid co-transporters. Pflugers Arch. 466: 155-72 Schiöth HB et al. (2013) Evolutionary origin of amino acid transporter families SLC32, SLC36 and
[PMID:24193407] SLC38 and physiological, pathological and therapeutic aspects. Mol. Aspects Med. 34: 571-85
Bröer S et al. (2011) The role of amino acid transporters in inherited and acquired diseases. Biochem. [PMID:23506890]
J. 436: 193-211 [PMID:21568940]
Comments: Zinc fluxes may be monitored through the use of radioisotopic Zn-65 or the fluorescent dye FluoZin 3.
The bicarbonate transport inhibitor DIDS has been reported to inhibit cation accumulation through ZIP14 [240].
Hojyo S et al. (2016) Zinc transporters and signaling in physiology and pathogenesis. Arch. Biochem. Kambe T et al. (2015) The Physiological, Biochemical, and Molecular Roles of Zinc Transporters in
Biophys. 611: 43-50 [PMID:27394923] Zinc Homeostasis and Metabolism. Physiol. Rev. 95: 749-784 [PMID:26084690]
Jeong J et al. (2013) The SLC39 family of zinc transporters. Mol. Aspects Med. 34: 612-9 Marger L et al. (2014) Zinc: an underappreciated modulatory factor of brain function. Biochem.
[PMID:23506894] Pharmacol. 91: 426-35 [PMID:25130547]
Kambe T et al. (2014) Current understanding of ZIP and ZnT zinc transporters in human health and
diseases. Cell. Mol. Life Sci. 71: 3281-95 [PMID:24710731]
Overview: Alongside the SLC11 family of proton-coupled metal [4, 140]. Ferroportin is essential for iron homeostasis [160]. Ferro- and degradation, lowering the levels of iron export to the blood.
transporters, ferroportin allows the accumulation of iron from the portin is expressed on the surface of cells that store and transport Novel therapeutic agents which stabilise ferroportin or protect it
diet. Whilst SLC11A2 functions on the apical membrane, ferro- iron, such as duodenal enterocytes, hepatocytes, adipocytes and from hepcidin-induced degradation are being developed as anti-
portin acts on the basolateral side of the enterocyte, as well as reticuloendothelial macrophages. Levels of ferroportin are regu- anemia agents. Anti-ferroportin monoclonal antibodies are such
regulating macrophage and placental iron levels. The predicted lated by its association with (binding to) hepcidin, a 25 amino an agent.
topology is of 12 TM domains, with intracellular termini [527], acid hormone responsive to circulating iron levels (amongst other
with the functional transporter potentially a dimeric arrangement signals). Hepcidin binding targets ferroportin for internalisation
Nomenclature Ferroportin
Systematic nomenclature SLC40A1
Common abbreviation IREG1
HGNC, UniProt SLC40A1, Q9NP59
Endogenous substrates Fe2+
Stoichiometry Unknown
Antibodies LY2928057 (Binding) [395]
Comments: Hepcidin (HAMP, P81172), cleaved into hepcidin-25 (HAMP, P81172) and hepcidin-20 (HAMP, P81172), is a small protein that increases upon inflammation, binds to ferroportin to regulate
its cellular distribution and degradation. Gene disruption in mice results in embryonic lethality [160], while loss-of-function mutations in man are associated with haemochromatosis [141].
McKie AT et al. (2004) The SLC40 basolateral iron transporter family (IREG1/ferroportin/MTP1). Montalbetti N et al. (2013) Mammalian iron transporters: families SLC11 and SLC40. Mol. Aspects
Pflugers Arch. 447: 801-6 [PMID:12836025] Med. 34: 270-87 [PMID:23506870]
Payandeh J et al. (2013) The structure and regulation of magnesium selective ion channels. Biochim. Schweigel-Röntgen M et al. (2014) SLC41 transporters–molecular identification and functional role.
Biophys. Acta 1828: 2778-92 [PMID:23954807] Curr Top Membr 73: 383-410 [PMID:24745990]
Sahni J et al. (2013) The SLC41 family of MgtE-like magnesium transporters. Mol. Aspects Med. 34:
620-8 [PMID:23506895]
Overview: Rhesus is commonly defined as a ‘factor’ that de- associates with these antigens and functions as an ammonium yeast, plants and bacteria. More recent evidence points to family
termines, in part, blood type, and whether neonates suffer from transporter. RhBG and RhBG are non-erythroid related sequences members being permeable to carbon dioxide, leading to the term
haemolytic disease of the newborn. These glycoprotein antigens associated with epithelia. Topological modelling suggests the pres- gas channels.
derive from two genes, RHCE (P18577) and RHD (Q02161), ex- ence of 12TM with cytoplasmic N- and C- termini. The majority
pressed on the surface of erythrocytes. On erythrocytes, RhAG of information on these transporters derives from orthologues in
Nomenclature Ammonium transporter Rh type A Ammonium transporter Rh type B Ammonium transporter Rh type C
Systematic nomenclature SLC42A1 SLC42A2 SLC42A3
Common abbreviation RhAG RhBG RhCG
HGNC, UniProt RHAG, Q02094 RHBG, Q9H310 RHCG, Q9UBD6
Substrates NH4 + [665], NH3 [528], CO2 [179] – NH3 [721]
Stoichiometry Unknown Unknown Unknown
Labelled ligands [14 C]methylamine (Binding) [286] – [14 C]methylamine (Binding) [423] – Mouse
Nakhoul NL et al. (2013) Characteristics of mammalian Rh glycoproteins (SLC42 transporters) and Weiner ID et al. (2014) Ammonia transport in the kidney by Rhesus glycoproteins. Am. J. Physiol.
their role in acid-base transport. Mol. Aspects Med. 34: 629-37 [PMID:23506896] Renal Physiol. 306: F1107-20 [PMID:24647713]
Weiner ID et al. (2011) Role of NH3 and NH4+ transporters in renal acid-base transport. Am. J.
Physiol. Renal Physiol. 300: F11-23 [PMID:21048022]
Overview: LAT3 (SLC43A1) and LAT4 (SLC43A2) are transporters put.ative TM domains with both N and C termini located intra- in the pancreas, liver, skeletal muscle and fetal liver [33] whereas
with system L amino acid transporter activity, along with the cellularly. They transport neutral amino acids in a manner inde- LAT4/SLC43A2 is primarily expressed in the placenta, kidney and
structurally and functionally distinct transporters LAT1 and LAT2 pendent of Na+ and Cl- and with two kinetic components [33, peripheral blood leukocytes [64]. SLC43A3 is expressed in vascular
that are members of the SLC7 family. LAT3 and LAT4 contain 12 64]. LAT3/SLC43A1 is expressed in human tissues at high levels endothelial cells [651] but remains to be characterised.
Searchable database: http://www.guidetopharmacology.org/index.jsp SLC43 family of large neutral amino acid transporters S473
Full Contents of ConciseGuide: http://onlinelibrary.wiley.com/doi/10.1111/bph.14753/full
S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: Transporters. British Journal of Pharmacology (2019) 176, S397–S493
Comments: Covalent modification of LAT3 by N-ethylmaleimide inhibits its function [33] and at LAT4 inhibits the low-, but not high-affinity component of transport [64].
Bodoy S et al. (2013) The small SLC43 family: facilitator system l amino acid transporters and the
orphan EEG1. Mol. Aspects Med. 34: 638-45 [PMID:23268354]
Overview: Members of the choline transporter-like family are en- with an affinity that is intermediate to that of the high affinity membranes [437]. Transport of choline by CTL2, which in rodents
coded by five genes (CTL1-CTL5) with further diversity occurring choline transporter CHT1 (SLC5A7) and the low affinity organic- is expressed as two isoforms (CTL2P1 and CLTP2; [370]) in lung,
through alternative splicing of CTL1, 4 and 5 [622]. CTL fam- cation transporters [OCT1 (SLC22A1) and OCT2 (SLC22A2)] [438]. colon, inner ear and spleen and to a lesser extent in brain, tongue,
ily members are putative 10TM domain proteins with extracel- CLT1 is expressed almost ubiquitously in human tissues [669] and liver, and kidney, has only recently been demonstrated [370, 458].
lular termini that mediate Na+ -independent transport of choline mediates choline transport across the plasma and mitochondrial CTL3-5 remain to be characterized functionally.
Comments: Data tabulated are features observed for CLT1 endogenous to: rat astrocytes [308]; rat renal tubule epithelial cells [683]; human colon carcinoma cells [373]; human keratinocytes [630] and
human neuroblastoma cells [684]. Choline uptake by CLT1 is inhibited by numerous organic cations (e.g. [308, 683, 684]). In the guinea-pig, CTL2 is a target for antibody-induced hearing loss [454] and
in man, a polymorphism in CTL2 constitutes the human neutrophil alloantigen-3a (HNA-3a; [256]).
Inazu M. (2014) Choline transporter-like proteins CTLs/SLC44 family as a novel molecular target for Traiffort E et al. (2013) The choline transporter-like family SLC44: properties and roles in human
cancer therapy. Biopharm Drug Dispos 35: 431-49 [PMID:24532461] diseases. Mol. Aspects Med. 34: 646-54 [PMID:23506897]
Bartölke R et al. (2014) Proton-associated sucrose transport of mammalian solute carrier family 45: Vitavska O et al. (2013) The SLC45 gene family of putative sugar transporters. Mol. Aspects Med. 34:
an analysis in Saccharomyces cerevisiae. Biochem. J. 464: 193-201 [PMID:25164149] 655-60 [PMID:23506898]
Hou Z et al. (2014) Biology of the major facilitative folate transporters SLC19A1 and SLC46A1. Curr Zhao R et al. (2011) Mechanisms of membrane transport of folates into cells and across epithelia.
Top Membr 73: 175-204 [PMID:24745983] Annu. Rev. Nutr. 31: 177-201 [PMID:21568705]
Matherly LH et al. (2014) The major facilitative folate transporters solute carrier 19A1 and solute Zhao R et al. (2013) Folate and thiamine transporters mediated by facilitative carriers (SLC19A1-3
carrier 46A1: biology and role in antifolate chemotherapy of cancer. Drug Metab. Dispos. 42: and SLC46A1) and folate receptors. Mol. Aspects Med. 34: 373-85 [PMID:23506878]
632-49 [PMID:24396145]
Wilson MR et al. (2015) Structural determinants of human proton-coupled folate transporter
oligomerization: role of GXXXG motifs and identification of oligomeric interfaces at transmem-
brane domains 3 and 6. Biochem. J. 469: 33-44 [PMID:25877470]
Comments: DAPI has been used to allow quantification of MATE1 and MATE2-mediated transport activity [693]. MATE2 and MATE2-B are inactive splice variants of MATE2-K [428].
Damme K et al. (2011) Mammalian MATE (SLC47A) transport proteins: impact on efflux of endoge- Wagner DJ et al. (2016) Polyspecific organic cation transporters and their impact on drug intracellular
nous substrates and xenobiotics. Drug Metab. Rev. 43: 499-523 [PMID:21923552] levels and pharmacodynamics. Pharmacol. Res. 111: 237-246 [PMID:27317943]
Motohashi H et al. (2013) Multidrug and toxin extrusion family SLC47: physiological, pharma- Yonezawa A et al. (2011) Importance of the multidrug and toxin extrusion MATE/SLC47A family to
cokinetic and toxicokinetic importance of MATE1 and MATE2-K. Mol. Aspects Med. 34: 661-8 pharmacokinetics, pharmacodynamics/toxicodynamics and pharmacogenomics. Br. J. Pharma-
[PMID:23506899] col. 164: 1817-25 [PMID:21457222]
Nies AT et al. (2016) Structure and function of multidrug and toxin extrusion proteins (MATEs)
and their relevance to drug therapy and personalized medicine. Arch. Toxicol. 90: 1555-84
[PMID:27165417]
Khan AA et al. (2013) Heme and FLVCR-related transporter families SLC48 and SLC49. Mol. Aspects
Med. 34: 669-82 [PMID:23506900]
Overview: FLVCR1 was initially identified as a cell-surface at- poiesis. Flvcr1 gene mutations have been identified as the cause Fowler’s syndrome, is associated with a loss-of-function mutation
tachment site for feline leukemia virus subgroup C [601], and of PCARP (posterior column ataxia with retinitis pigmentosa in FLVCR2 [435].
later identified as a cell surface accumulation which exports heme (PCARP) [516].There are three paralogs of FLVCR1 in the human
from the cytosol [513]. A recent study indicates that an isoform of genome.
FLVCR1 is located in the mitochondria, the site of the final steps FLVCR2, most similar to FLVCR1 [403], has been reported to func- The functions of the other two members of the SLC49 family,
of heme synthesis, and appears to transport heme into the cytosol tion as a heme importer [163]. In addition, a congenital syndrome MFSD7 and DIRC2, are unknown, although DIRC2 has been im-
[113]. FLVCR-mediated heme transport is essential for erythro- of proliferative vasculopathy and hydranencephaly, also known as plicated in hereditary renal carcinomas [63].
Nomenclature Feline leukemia virus subgroup C cellular receptor family, member 1 Feline leukemia virus subgroup C cellular receptor family, member 2
Systematic nomenclature SLC49A1 SLC49A2
Common abbreviation FLVCR1 FLVCR2
HGNC, UniProt FLVCR1, Q9Y5Y0 FLVCR2, Q9UPI3
Substrates heme [513] heme [163]
Stoichiometry Unknown Unknown
Comments: Non-functional splice alternatives of FLVCR1 have been implicated as a cause of a congenital red cell aplasia, Diamond Blackfan anemia [525].
Khan AA et al. (2013) Heme and FLVCR-related transporter families SLC48 and SLC49. Mol. Aspects Khan AA et al. (2011) Control of intracellular heme levels: heme transporters and heme oxygenases.
Med. 34: 669-82 [PMID:23506900] Biochim. Biophys. Acta 1813: 668-82 [PMID:21238504]
Wright EM. (2013) Glucose transport families SLC5 and SLC50. Mol. Aspects Med. 34: 183-96 Wright EM et al. (2011) Biology of human sodium glucose transporters. Physiol. Rev. 91: 733-94
[PMID:23506865] [PMID:21527736]
Overview: The SLC51 organic solute transporter family of trans- pendent of sodium, potassium, chloride ions or protons [41, tein, while OSTβ is a single TM ’ancillary’ protein, both of which
porters is a pair of heterodimeric proteins which regulate bile 137]. OSTα/OSTβ heterodimers have been shown to trans- are thought to have intracellular C-termini [400]. Both pro-
salt movements in the small intestine, bile duct, and liver, as port [3 H]taurocholic acid, [3 H]dehydroepiandrosterone sulphate, teins function in solute transport and bimolecular fluorescence
part of the enterohepatic circulation [41, 137]. OSTα/OSTβ is [3 H]estrone-3-sulphate, [3 H]pregnenolone sulphate and complementation studies suggest the possibility of OSTα homo-
also expressed in steroidogenic cells of the brain and adrenal 3
[ H]dehydroepiandrosterone sulphate [41, 137, 193]. oligomers, as well as OSTα/OSTβ hetero-oligomers [117, 400]. An
gland, where it may contribute to steroid movement [193]. OSTα/OSTβ-mediated transport of bile salts is inhibited by inherited mutation in OSTβ is associated with congenital diarrhea
Bile acid transport is suggested to be facilitative and inde- clofazimine [635]. OSTα is suggested to be a seven TM pro- in children [591].
Ballatori N. (2011) Pleiotropic functions of the organic solute transporter Ostα-Ostβ. Dig Dis 29: 13-7 Dawson PA. (2011) Role of the intestinal bile acid transporters in bile acid and drug disposition.
[PMID:21691099] Handb Exp Pharmacol 169-203 [PMID:21103970]
Ballatori N et al. (2013) The heteromeric organic solute transporter, OSTα-OSTβ/SLC51: a transporter
for steroid-derived molecules. Mol. Aspects Med. 34: 683-92 [PMID:23506901]
Nomenclature solute carrier family 52 member 1 solute carrier family 52 member 2 solute carrier family 52 member 3
Systematic nomenclature SLC52A1 SLC52A2 SLC52A3
Common abbreviation RFVT1 RFVT2 RFVT3
HGNC, UniProt SLC52A1, Q9NWF4 SLC52A2, Q9HAB3 SLC52A3, Q9NQ40
Endogenous substrates riboflavin (Km 1.3×10−3 M) [692] riboflavin (Km 9.8×10−4 M) [692] riboflavin (Km 3.3×10−4 M) [692]
Stoichiometry Unknown Unknown H+ -dependent
Comments: Although expressed elsewhere, RFVT3 is found on the luminal surface of intestinal epithelium and is thought to mediate uptake of dietary riboflavin, while RFVT1 and RFVT2 are thought
to allow movement from the epithelium into the blood.
Yonezawa A et al. (2013) Novel riboflavin transporter family RFVT/SLC52: identification, nomen-
clature, functional characterization and genetic diseases of RFVT/SLC52. Mol. Aspects Med. 34:
693-701 [PMID:23506902]
Nomenclature mitochondrial pyruvate carrier 1 mitochondrial pyruvate carrier 2 mitochondrial pyruvate carrier 1 like
Systematic nomenclature SLC54A1 SLC54A2 SLC54A3
HGNC, UniProt MPC1, Q9Y5U8 MPC2, O95563 MPC1L, P0DKB6
Substrates Pyruvate [73, 289] Pyruvate [73] Pyruvate [642]
Comments SLC54A1 is ubiquitously expressed [642]. SLC54A2 is ubiquitously expressed [642]. SLC54A3 is expressed in testis, postmeiotic spermatids and
sperm cells [642].
Comments: SLC54 family transporters appear to function as mechanisms for accumulating pyruvate into mitochondria to link glycolysis with oxidative phosphorylation.
Nomenclature leucine zipper and EF-hand containing transmembrane leucine zipper and EF-hand containing transmembrane LETM1 domain containing 1
protein 1 protein 2
Systematic nomenclature SLC55A1 SLC55A2 SLC55A3
HGNC, UniProt LETM1, O95202 LETM2, Q2VYF4 LETMD1, Q6P1Q0
Transport type Exchanger / Ca2+ :H+ [328, 569] – –
Exchanger / K+ :H+ [154, 468]
Substrates Ca2+ , K+ , H+ [154, 468, 469, 725] – –
Comments SLC55A1 is ubiquitously expressed [178]. Arguments – –
against SLC55A1’s role as a Ca2+ transporter are outlined
by Zotova et al. (2010) [725].
Comments: The family of SLC55 mitochondrial transporters appear to regulate ion fluxes and to maintain tubular networks.
SLC56 Sideroflexins
Transporters → SLC superfamily of solute carriers → SLC56 Sideroflexins
Nomenclature NIPA magnesium transporter 1 NIPA magnesium transporter 2 NIPA like domain containing 1 NIPA like domain containing 3
Systematic nomenclature SLC57A1 SLC57A2 SLC57A3 SLC57A5
HGNC, UniProt NIPA1, Q7RTP0 NIPA2, Q8N8Q9 NIPAL1, Q6NVV3 NIPAL3, Q6P499
Substrates Sr2+ , Fe2+ and Co2+ to a lesser extent [250], Mg2+ [250] Mg2+ , Sr2+ , Ba2+ , Fe2+ , Cu2+ [250] –
Mg2+ [249]
Comments Human tissue expression: Constitutively – – –
expressed at low levels, with significant
enrichment in the brain [515].
Mouse tissue expression: Widely expressed,
including in the heart, kidney, liver, colon,
less in the brain, and not in the small
intestine [249].
Nomenclature major facilitator superfamily domain containing 2A major facilitator superfamily domain containing 2B
Systematic nomenclature SLC59A1 SLC59A2
HGNC, UniProt MFSD2A, Q8NA29 MFSD2B, A6NFX1
Transport type Co-transporter: LPC:Na+ , uptake –
Substrates LPC (lysophosphatidylcholine) form of DHA (docosahexaenoic acid) [464] –
Comments MFSD2B/SLC59A has been suggested to be a sphingosine 1-phosphate transporter in Expressed in the spleen, lung, testis and subcellularly in the ER [19].
erythropoietic cells [364]. It is expressed in brain, intestine, kidney, liver, lung, mammary
gland, and prostate [19]; relatively low expression in BAT (brown adipose tissue), but
upregulated during cold-induced thermogenesis [19]. Subcellular locations: plasma
membrane [656] and ER [19].
Nomenclature major facilitator superfamily domain containing 4A major facilitator superfamily domain containing 4B
Systematic nomenclature SLC60A1 SLC60A2
HGNC, UniProt MFSD4A, Q8N468 MFSD4B, Q5TF39
Transport type – Co-transporter / Na+ (1:1) uptake (Rat) [295]
Substrates – α-Me-glucose, D-glucose [295]
Inhibitors – phloretin [295] – Rat, phlorizin [295] – Rat, urea [460] – Rat
Comments – Expressed in rat kidney (cortex and medulla), brain, liver and lung [295].
Nomenclature NPC intracellular cholesterol transporter 1 NPC1 like intracellular cholesterol transporter 1
Systematic nomenclature SLC65A1 SLC65A2
HGNC, UniProt NPC1, O15118 NPC1L1, Q9UHC9
Substrates Cholesterol [309, 310, 496] Cholesterol [10]
Selective antagonists – ezetimibe (Inhibition) (pKd 6.7) [227]
Comments Expression is ubiquitous [10], with highest levels detected in liver, lung, and pancreas Expressed in small intestine, gallbladder, liver, testis and stomach [10].
[136]. NPC1 plays a critical role in the regulation of intracellular cholesterol trafficking
[93]. Mutations in the NPC1 gene have been identified in patients with the lipid storage
disorder Niemann-Pick disease type C1 [62, 93, 255, 686].
Overview: The SLCO superfamily is comprised of the organic an- cells throughout the body. They have 12 TM domains and intra- the multispecificity of these proteins, this guide lists classes of sub-
ion transporting polypeptides (OATPs). The 11 human OATPs are cellular termini, with multiple putative glycosylation sites. OATPs strates and inhibitors for each family member. More comprehen-
divided into 6 families and ten subfamilies based on amino acid mediate the sodium-independent uptake of a wide range of am- sive lists of substrates, inhibitors, and their relative affinities may
identity. These proteins are located on the plasma membrane of phiphilic substrates, including many drugs and toxins. Due to be found in the review articles listed below.
Searchable database: http://www.guidetopharmacology.org/index.jsp SLCO family of organic anion transporting polypeptides S488
Full Contents of ConciseGuide: http://onlinelibrary.wiley.com/doi/10.1111/bph.14753/full
S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: Transporters. British Journal of Pharmacology (2019) 176, S397–S493
Hagenbuch B et al. (2013) The SLCO (former SLC21) superfamily of transporters. Mol. Aspects Med. Roth M et al. (2012) OATPs, OATs and OCTs: the organic anion and cation transporters of the SLCO
34: 396-412 [PMID:23506880] and SLC22A gene superfamilies. Br. J. Pharmacol. 165: 1260-87 [PMID:22013971]
Hillgren KM et al. (2013) Emerging transporters of clinical importance: an update from the Interna- Zamek-Gliszczynski MJ et al. (2018) Transporters in Drug Development: 2018 ITC Recommenda-
tional Transporter Consortium. Clin. Pharmacol. Ther. 94: 52-63 [PMID:23588305] tions for Transporters of Emerging Clinical Importance. Clin. Pharmacol. Ther. 104: 890-899
International Transporter Consortium et al. (2010) Membrane transporters in drug development. [PMID:30091177]
Nat Rev Drug Discov 9: 215-36 [PMID:20190787]
Lee HH et al. (2017) Interindividual and interethnic variability in drug disposition: polymorphisms
in organic anion transporting polypeptide 1B1 (OATP1B1; SLCO1B1). Br J Clin Pharmacol 83:
1176-1184 [PMID:27936281]
Bhutia YD et al. (2016) SLC transporters as a novel class of tumour suppressors: identity, function Neul C et al. (2016) Impact of Membrane Drug Transporters on Resistance to Small-Molecule Tyrosine
and molecular mechanisms. Biochem. J. 473: 1113-24 [PMID:27118869] Kinase Inhibitors. Trends Pharmacol. Sci. 37: 904-932 [PMID:27659854]
Colas C et al. (2016) SLC Transporters: Structure, Function, and Drug Discovery. Medchemcomm 7: Nigam SK. (2015) What do drug transporters really do? Nat Rev Drug Discov 14: 29-44
1069-1081 [PMID:27672436] [PMID:25475361]
César-Razquin A et al. (2015) A Call for Systematic Research on Solute Carriers. Cell 162: 478-87 Pedersen NB et al. (2016) Glycosylation of solute carriers: mechanisms and functional consequences.
[PMID:26232220] Pflugers Arch. 468: 159-76 [PMID:26383868]
Lin L et al. (2015) SLC transporters as therapeutic targets: emerging opportunities. Nat Rev Drug Perland E et al. (2017) Classification Systems of Secondary Active Transporters. Trends Pharmacol. Sci.
Discov 14: 543-60 [PMID:26111766] 38: 305-315 [PMID:27939446]
Naª¦cz KA. (2017) Solute Carriers in the Blood-Brain Barier: Safety in Abundance. Neurochem. Res. Rives ML et al. (2017) Potentiating SLC transporter activity: Emerging drug discovery opportunities.
42: 795-809 [PMID:27503090] Biochem. Pharmacol. 135: 1-11 [PMID:28214518]
Searchable database: http://www.guidetopharmacology.org/index.jsp SLCO family of organic anion transporting polypeptides S489
Full Contents of ConciseGuide: http://onlinelibrary.wiley.com/doi/10.1111/bph.14753/full
S.P.H. Alexander et al. The Concise Guide to PHARMACOLOGY 2019/20: Transporters. British Journal of Pharmacology (2019) 176, S397–S493
References
1. Abbot EL et al. (2006) [16331283] 46. Beart PM et al. (2007) [17088867] 93. Carstea ED et al. (1997) [9211849] 136. Davies JP et al. (2000) [10783261]
2. Abramson J et al. (2009) [19631523] 47. Belanger AM et al. (2018) [30046012] 94. Caulfield MJ et al. (2008) [18842065] 137. Dawson PA et al. (2005) [15563450]
3. Agu R et al. (2011) [21366347] 48. Bellocchio EE et al. (2000) [10938000] 95. Caulfield WL et al. (2001) [11495577] 138. Dawson PA et al. (2009) [19498215]
4. Aguirre P et al. (2005) [15667655] 49. Ben-Daniel R et al. (2008) [18487050] 96. Cerveny L et al. (2018) [30097436] 139. de Carvalho FD et al. (2011) [20980265]
5. Agulhon C et al. (2003) [12761825] 50. Bergeron R et al. (1998) [9861038] 97. Cha SH et al. (2000) [10660625] 140. De Domenico I et al. (2007) [17077321]
6. Akazawa T et al. (2018) [30135242] 51. Betz H et al. (2006) [16417482] 98. Chan BS et al. (1998) [9506966] 141. De Domenico I et al. (2005) [15956209]
7. Albers A et al. (2001) [11692272] 52. Bhardwaj RK et al. (2006) [16289537] 99. Chang MH et al. (2009) [19365592] 142. Dean M et al. (2001) [11441126]
8. Albrecht C et al. (2007) [16586097] 53. Bhardwaj RK et al. (2005) [15901802] 100. Chao EC et al. (2010) [20508640] 143. Delpire E et al. (2009) [19279215]
9. Almqvist J et al. (2004) [15260472] 54. Bhat BG et al. (2003) [12810816] 101. Charkoudian LK et al. (2012) Natural 144. Demaegd D et al. (2013) [23569283]
10. Altmann SW et al. (2004) [14976318] 55. Bianchi J et al. (1986) [3945643] product inhibitors of glucose-6-phosphate 145. Demirel Ö et al. (2012) [22641697]
11. Amara SG et al. (1993) [8103691] 56. Biegel A et al. (2005) [15974593] translocase. MedChemComm. 3: 926-931 146. Dennis M et al. (2013) Compositions and
12. Anand BS et al. (2003) [12538834] 57. Biegel A et al. (2006) [16868651] 102. Charrier L et al. (2006) [16568107] methods for the diagnosis and treatment of
13. Anderson CM et al. (2008) [18599538] 58. Bissonnette P et al. (2004) [15181167] 103. Chavan H et al. (2015) [25623066] tumor. Patent number: US8535675 B2.
14. Anderson CM et al. (2004) [15521011] 59. Bizhanova A et al. (2009) [19196800] 104. Cheeseman C. (2008) [18477702] 147. DeStefano GM et al. (2014) [24831815]
15. Anderson CM et al. (2009) [19074966] 60. Blackburn C et al. (2006) [16644217] 105. Chen LQ et al. (2010) [21107422] 148. Dhar TG et al. (1994) [8057281]
16. Anderson CM et al. (2010) [19789362] 61. Blair BG et al. (2009) [19509135] 106. Chen NH et al. (2004) [12719981] 149. Dhar TGM et al. (1996) On the bioactive
17. Anderson CM et al. (2005) [15754324] 62. Blom TS et al. (2003) [12554680] 107. Chen SY et al. (2008) [18337460] conformation of the GABA uptake inhibitor
18. Andrini O et al. (2008) [18989094] 63. Bodmer D et al. (2002) [11912179] 108. Chen Y et al. (2007) [17495125] SK&F 89976-A. Bioorganic and medicinal
19. Angers M et al. (2008) [18694395] 64. Bodoy S et al. (2005) [15659399] 109. Chen Z et al. (2003) [12727219] chemistry letters 6: 1535 -1540
20. Ansermet C et al. (2017) [27799484] 65. Borden LA et al. (1994) [7874447] 110. Chen ZQ et al. (2006) [16421098] 150. Di Daniel E et al. (2009) [19607714]
21. Aouameur R et al. (2007) [17932225] 66. Borden LA et al. (1994) [7851497] 111. Chen ZS et al. (1999) [10570049] 151. Diaz GA et al. (1999) [10391223]
22. Apparsundaram S et al. (2000) [11027560] 67. Borst P et al. (2007) [16586096] 112. Cheng Q et al. (2017) [28176326] 152. Dieck ST et al. (1999) [9888294]
23. Apricò K et al. (2004) [14994336] 68. Boudker O et al. (2007) [17230192] 113. Chiabrando D et al. (2012) [23187127] 153. Diez-Sampedro A et al. (2003) [13130073]
24. Apricò K et al. (2007) [17590480] 69. Boulay D et al. (2008) [18621075] 114. Choi MK. (2012) [22644860] 154. Dimmer KS et al. (2008) [17925330]
25. Apricó K et al. (2001) [11389172] 70. Bourgeois F et al. (2005) [15613375] 115. Choi MK et al. (2015) [25011570] 155. Dodd JR et al. (2007) [17400549]
26. Armstrong D et al. (2014) [24414167] 71. Bravo DT et al. (2005) [15979764] 116. Chong X et al. (1992) [1417961] 156. Doege H et al. (2001) [11583593]
27. Arriza JL et al. (1993) [8101838] 72. Bravo DT et al. (2004) [15485505] 117. Christian WV et al. (2012) [22535958] 157. Dohán O et al. (2007) [18077370]
28. Ashikov A et al. (2005) [15911612] 73. Bricker DK et al. (2012) [22628558] 118. Chu XY et al. (2001) [11602669] 158. Dong H et al. (2002) [11916852]
29. Assaraf YG et al. (1998) [9525913] 74. Brown A et al. (2001) [11454468] 119. Clausen RP et al. (2006) [17175818] 159. Dong Z et al. (2013) [23339484]
30. Aubrey KR et al. (2000) [10860934] 75. Bröer A et al. (2002) [11850497] 120. Coady MJ et al. (2007) [17526579] 160. Donovan A et al. (2005) [16054062]
31. Auerbach SS et al. National Toxicology 76. Bröer A et al. (2009) [19657969] 121. Coady MJ et al. (2002) [12133831] 161. Dorwart MR et al. (2007) [17673510]
Program: Dept of Health and Human 77. Bröer A et al. (1999) [10537079] 122. Coelho D et al. (2012) [22922874] 162. Draoui N et al. (2013) [24095010]
Services. 78. Bröer A et al. (2006) [16185194] 123. Cohen-Kfir E et al. (2005) [15829583] 163. Duffy SP et al. (2010) [20823265]
32. Azzi A et al. (1993) [8132491] 79. Bröer A et al. (2000) [10698697] 124. Colleoni S et al. (2008) [18451317] 164. Dunlop J. (2006) [16368269]
33. Babu E et al. (2003) [12930836] 80. Bröer S. (2006) [16540203] 125. Colton CK et al. (2010) [20508255] 165. Dunlop J et al. (2006) [17017964]
34. Bagrov AY et al. (2009) [19325075] 81. Bröer S. (2008) [18400692] 126. Coon et al. (2004) Abstract Viewer/Itinerary 166. Dunlop J et al. (2003) [14517179]
35. Bailey CG et al. (2011) [21123949] 82. Bröer S et al. (2008) [19033659] Planner, Program No. 168.10. Society 167. Dunlop J et al. (2005) [16014807]
36. Bailey DG et al. (2007) [17301733] 83. Burant CF et al. (1992) [1634504] for Neuroscience: 168. Dutta B et al. (1999) [10542220]
37. Bakos E et al. (2007) [17187268] 84. Burger S et al. (2011) [21742018] 127. Counillon L et al. (1993) [8246907] 169. Döring F et al. (1998) [9637710]
38. Baldwin SA et al. (2005) [15701636] 85. Burns CM et al. (2011) [20719377] 128. Covitz KM et al. (1996) [8956326] 170. Edington AR et al. (2009) [19875446]
39. Balimane P et al. (2000) [11180195] 86. Busch AE et al. (1996) [8643577] 129. Craddock AL et al. (1998) [9458785] 171. Edwards N et al. (2011) [20691150]
40. Balimane PV et al. (1998) [9753615] 87. Buyse M et al. (2001) [11714740] 130. Cuboni S et al. (2014) [25318072] 172. Edwards N et al. (2018) [29058016]
41. Ballatori N et al. (2005) [16317684] 88. Buyse M et al. (2003) [14578196] 131. Dai W et al. (1999) [9882430] 173. Efange SM et al. (1995) [7702637]
42. Banerjee A et al. (2006) [16411770] 89. Byrne JA et al. (2002) [12404239] 132. Dalton TP et al. (2005) [15722412] 174. Eiden LE et al. (2004) [12827358]
43. Barnes K et al. (2006) [16873718] 90. Böhmer C et al. (2005) [15804236] 133. Daniels G et al. (2015) [25896650] 175. Eiden LE et al. (2011) [21272013]
44. Battini JL et al. (1999) [9990033] 91. Carland JE et al. (2013) [22978602] 134. Danthi SJ et al. (2019) [30589598] 176. Eliasof S et al. (2001) [11299317]
45. Bayeva M et al. (2013) [23720443] 92. Carr G et al. (2009) [19910700] 135. Darcel NP et al. (2005) [15930458] 177. Elliott AM et al. (2009) [19147539]
178. Endele S et al. (1999) [10486213] 228. Garzel B et al. (2014) [24335466] 270. Guo A et al. (1999) [10087037] 320. Ito S et al. (2010) [20065018]
179. Endeward V et al. (2008) [17712059] 229. Gasnier B. (2004) [12750892] 271. Gupta N et al. (2006) [16375929] 321. Ito Y et al. (2001) [11527541]
180. Engel K et al. (2005) [16099839] 230. Gasnier B. (2000) [10865121] 272. Gupte A et al. (2009) [19097778] 322. Iwamoto H et al. (2006) [17005849]
181. Enomoto A et al. (2002) [12024214] 231. Gebhardt FM et al. (2010) [20688910] 273. Hager K et al. (1995) [7537337] 323. Iwamoto T et al. (2006) [16973719]
182. Erickson JD et al. (1993) [8245983] 232. Gendreau S et al. (2004) [15265858] 274. Hallén S et al. (1999) [10471288] 324. Iwao T et al. (2014) [23822979]
183. Erickson JD et al. (1996) [8643547] 233. Gengo PJ et al. (2005) Monoaminergic 275. Hamada T et al. (2008) [18670416] 325. Jappar D et al. (2010) [20660104]
184. Erickson JD et al. (1994) [8071310] transporter binding and inhibition profile of 276. Hammond JR. (2000) [10763851] 326. Jensen AA et al. (2009) [19161278]
185. Eskandari S et al. (1997) [9341168] dapoxetine, a medication for the treatment 277. Hammond JR et al. (2004) [14634039] 327. Jeong HJ et al. (2010) [20860669]
186. Esslinger CS et al. (2005) [16183084] of premature ejaculation [abstract]. J Urol 278. Han H et al. (1998) [9706043] 328. Jiang D et al. (2009) [19797662]
187. Esslinger CS et al. (2005) [15670919] 173: Abstract 878 279. Han X et al. (2006) [16734743] 329. Jiang J et al. (2011) [20708631]
188. Etoga JL et al. (2010) [20303751] 234. Geyer J et al. (2007) [17491011] 280. Hannaert P et al. (2002) [11882915] 330. Jonas MC et al. (2010) [20826464]
189. Eulenburg V et al. (2005) [15950877] 235. Geyer J et al. (2008) [18355966] 281. Harvey RJ et al. (2008) [18707791] 331. Jost N et al. (2013) [23647096]
190. Faergeman NJ et al. (1997) [9079682] 236. Geyer J et al. (2004) [15020217] 282. Hatanaka T et al. (2001) [11342143] 332. Ju P et al. (2004) [15031290]
191. Fan SJ et al. (2018) [29971004] 237. Gillberg P-G et al. (2013) Ibat inhibitors 283. Hatanaka T et al. (2000) [10930503] 333. Juge N et al. (2010) [20920794]
192. Fan SJ et al. (2016) [26434594] for the treatment of liver diseases Patent 284. Hatanaka T et al. (2001) [11306607] 334. Juge N et al. (2009) [19843525]
193. Fang F et al. (2010) [20649839] number: US20130225511A1. 285. Helias V et al. (2012) [22246506] 335. Kamiyama S et al. (2006) [16492677]
194. Fattorini G et al. (2009) [19627441] 238. Gimeno RE et al. (2003) [12556534] 286. Hemker MB et al. (2003) [12846905] 336. Kamiyama S et al. (2003) [12716889]
195. Favari E et al. (2004) [15514211] 239. Giovannini D et al. (2013) [23791524] 287. Herdon HJ et al. (2010) [20691713] 337. Kanai N et al. (1995) [7754369]
196. Fehrenbach T et al. (2003) [14530907] 240. Girijashanker K et al. (2008) [18270315] 288. Herrera-Ruiz D et al. (2004) [15832510] 338. Kanai Y et al. (2004) [14530974]
197. Fei YJ et al. (2000) [10823827] 241. Godoy JR et al. (2007) [17628207] 289. Herzig S et al. (2012) [22628554] 339. Kanai Y et al. (2003) [14612154]
198. Ferdinandusse S et al. (2015) [25168382] 242. Gomeza J et al. (2006) [16722246] 290. Hiasa M et al. (2014) [25355561] 340. Kanai Y et al. (1994) [8282810]
199. Ferguson SM et al. (2004) [15173594] 243. Gomeza J et al. (2003) [14622582] 291. Hirschfield GM et al. (2013) [23583734] 341. Kanamori A et al. (1997) [9096318]
200. Ferguson SM et al. (2004) [14993474] 244. Gomeza J et al. (2003) [14622583] 292. Ho AM et al. (2000) [10894769] 342. Kang N et al. (2010) [20595384]
201. Fernandes CF et al. (2007) [17632081] 245. Gong Y et al. (2017) [28943923] 293. Ho HT et al. (2011) [21816955] 343. Kang SY et al. (2010) [20637636]
202. Fiermonte G et al. (2003) [12807890] 246. Gopal E et al. (2005) [15651982] 294. Hollingworth P et al. (2011) [21460840] 344. Karlgren M et al. (2012) [22541068]
203. Fiermonte G et al. (2009) [19429682] 247. Gorboulev V et al. (1997) [9260930] 295. Horiba N et al. (2003) [12590146] 345. Karunakaran S et al. (2008) [18522536]
204. Fischer WJ et al. (2005) [15737679] 248. Goursaud S et al. (2011) [21730107] 296. Hsu CL et al. (2012) [22174130] 346. Kato Y et al. (2017) Identification of a vesic-
205. Fleming MD et al. (2001) [11274051] 249. Goytain A et al. (2007) [17166836] 297. Hu Y et al. (2018) [29784761] ular ATP release inhibitor for the treatment
206. Foltz M et al. (2004) [15345686] 250. Goytain A et al. (2008) [18667602] 298. Hu Y et al. (2014) [24548120] of neuropathic and inflammatory pain Pro-
207. Fontana AC et al. (2007) [17646426] 251. Goytain A et al. (2005) [15804357] 299. Hu Z et al. (2000) [10615129] ceedings of the National Academy of Sciences of
208. Fontana AC et al. (2003) [12890709] 252. Goytain A et al. (2005) [15713785] 300. Huang S et al. (2009) [19074430] the United States of America
209. Forrest LR et al. (2009) [19996368] 253. Goytain A et al. (2005) [15809054] 301. Hummel CS et al. (2011) [20980548] 347. Kemp S et al. (2016) [27312864]
210. Forster IC et al. (1999) [10198426] 254. Graf J et al. (2005) [15714523] 302. Hägglund MG et al. (2011) [21511949] 348. Kemp S et al. (2011) [21488864]
211. Foulquier F et al. (2012) [22683087] 255. Greer WL et al. (1999) [10521290] 303. Ibberson M et al. (2000) [10671487] 349. Kenda BM et al. (2004) [14736235]
212. Fredriksson R et al. (2008) [18948099] 256. Greinacher A et al. (2010) [20037594] 304. Ichida K et al. (2003) [12472777] 350. Kennedy DJ et al. (2005) [15644866]
213. Friesema EC et al. (2006) [16887882] 257. Grewer C et al. (2005) [16128593] 305. Ichikawa Y et al. (2012) [22375032] 351. Kerr ID et al. (2011) [21175590]
214. Froimowitz M et al. (2007) [17228864] 258. Grewer C et al. (2004) [15107471] 306. Iharada M et al. (2010) [20566650] 352. Khare P et al. (2010) [20225888]
215. Fu Y et al. (2013) [23931754] 259. Grewer C et al. (2005) [15834685] 307. Inagaki N et al. (1995) [7502040] 353. Kim K et al. (2011) [21792905]
216. Fujinami K et al. (2015) [25312043] 260. Groneberg DA et al. (2001) [11518682] 308. Inazu M et al. (2005) [16000150] 354. Kim KH et al. (2005) [15591059]
217. Fujita T et al. (2004) [14715149] 261. Grozio A et al. (2019) Slc12a8 is a nicoti- 309. Infante RE et al. (2008) [17989073] 355. Kim RB et al. (1999) [10565843]
218. Fülep GH et al. (2006) [16766089] namide mononucleotide transporter Nature 310. Infante RE et al. (2008) [17989072] 356. Kimura H et al. (2002) [11907186]
219. Gabernet L et al. (2005) [15555781] Metabolism 1: 47˘57 311. Inoue T et al. (2017) [28410751] 357. Klaassen CD et al. (2010) [20103563]
220. Gameiro A et al. (2011) [21641307] 262. Grunewald M et al. (2000) [10734120] 312. Irie M et al. (2001) [11454935] 358. Klitgaard H et al. (2007) [23484603]
221. Ganapathy ME et al. (1995) [7592745] 263. Gründemann D et al. (1999) [10385678] 313. Ishida N et al. (1998) [9644260] 359. Knutsen LJ et al. (1999) [10479278]
222. Ganapathy ME et al. (1998) [9610386] 264. Gründemann D et al. (1998) [10196521] 314. Ishida N et al. (2005) [15607426] 360. Knütter I et al. (2004) [14706812]
223. Ganapathy ME et al. (1997) [9092716] 265. Gu H et al. (1994) [8125921] 315. Ishida N et al. (1996) [9010752] 361. Knütter I et al. (2009) [18824524]
224. Ganapathy V et al. (2008) [18446519] 266. Gu HH et al. (1996) [8636118] 316. Ishida N et al. (1999) [10393322] 362. Knütter I et al. (2001) [11284702]
225. Ganapathy V et al. (2009) [18992769] 267. Gui C et al. (2010) [20448812] 317. Ishida S et al. (2002) [12370430] 363. Knütter I et al. (2008) [18713951]
226. Ganel R et al. (2006) [16274998] 268. Guile SD et al. (2006) [16455256] 318. Ismair MG et al. (2006) [17487240] 364. Kobayashi N et al. (2018) [29563527]
227. Garcia-Calvo M et al. (2005) [15928087] 269. Gunshin H et al. (1997) [9242408] 319. Itagaki S et al. (2006) [16729224] 365. Kobayashi T et al. (2014) [25238095]
366. Koch HP et al. (2007) [17360917] 414. Lytton J et al. (1991) [1832668] 464. Nguyen LN et al. (2014) [24828044] 514. Raffel DM et al. (2004) [15300361]
367. Koch HP et al. (1999) [10570036] 415. Löscher W et al. (2016) [27752944] 465. Nicolas JM et al. (2016) [26663401] 515. Rainier S et al. (2003) [14508710]
368. Koepsell H. (2013) [23506881] 416. Lühn K et al. (2001) [11326279] 466. Nigam SK et al. (2018) [29847376] 516. Rajadhyaksha AM et al. (2010) [21070897]
369. Kolisek M et al. (2012) [22031603] 417. MacDonald L et al. (2002) [11895172] 467. Nothmann D et al. (2011) [21127051] 517. Rajagopal A et al. (2008) [18418376]
370. Kommareddi PK et al. (2010) [20665236] 418. MacGrogan D et al. (1996) [8661104] 468. Nowikovsky K et al. (2004) [15138253] 518. Rajgopal A et al. (2001) [11731220]
371. Kory N et al. (2018) [30442778] 419. Machtens JP et al. (2011) [21572047] 469. Nowikovsky K et al. (2007) [17541427] 519. Ravera S et al. (2007) [17494632]
372. Kottra G et al. (2013) [24744852] 420. Maciver B et al. (2008) [18256317] 470. Noyer M et al. (1995) [8605950] 520. Reddy VS et al. (2012) [22458847]
373. Kouji H et al. (2009) [19135976] 421. Madeo M et al. (2014) [25326386] 471. Noé J et al. (2007) [17470528] 521. Rees EM et al. (2004) [15494390]
374. Krishnamurthy PC et al. (2006) [17006453] 422. Madsen KK et al. (2010) [20026354] 472. Numata M et al. (2001) [11279194] 522. Rehmann H. (2012) [22260657]
375. Krishnaswamy A et al. (2009) [19186169] 423. Mak DO et al. (2006) [16131648] 473. Núñez E et al. (2000) [10694221] 523. Reid G et al. (2003) [12835412]
376. Kristensen AS et al. (2011) [21752877] 424. Mallorga PJ et al. (2003) [12941372] 474. O’Callaghan KM et al. (2010) [19875448] 524. Reith ME et al. (1996) [8878059]
377. Ksander BR et al. (2014) [25030174] 425. Mandal A et al. (2016) [27543355] 475. Ocheltree SM et al. (2004) [14600253] 525. Rey MA et al. (2008) [18815190]
378. Kung MP et al. (1994) [7855735] 426. Manolescu AR et al. (2007) [17710649] 476. Oda K et al. (2010) [19900191] 526. Reyes N et al. (2009) [19924125]
379. Kusuhara H et al. (1999) [10224140] 427. Martens H et al. (2008) [19052203] 477. Ohta KY et al. (2006) [16928787] 527. Rice AE et al. (2009) [19150361]
380. Kvist T et al. (2009) [19275529] 428. Masuda S et al. (2006) [16807400] 478. Okuda T et al. (2003) [12675135] 528. Ripoche P et al. (2004) [15572441]
381. Lapinsky DJ et al. (2011) [21129986] 429. McIntire SL et al. (1997) [9349821] 479. Okuda T et al. (2000) [11068039] 529. Rodriguez L et al. (2006) [16632403]
382. Larráyoz IM et al. (2006) [16837649] 430. Meier PJ et al. (1997) [9398014] 480. Oppedisano F et al. (2010) [20599776] 530. Rogers S et al. (2003) [12914765]
383. Larsen M et al. (2009) [19594759] 431. Meredith D et al. (1998) [9882198] 481. Ordovás L et al. (2006) [17065791] 531. Romano A et al. (2010) [19913073]
384. Lau CL et al. (2011) [21309758] 432. Merlin D et al. (2001) [11375948] 482. Otsuka M et al. (2005) [16330770] 532. Romera C et al. (2007) [17213861]
385. Leary GP et al. (2007) [17360916] 433. Merlin D et al. (1998) [9835627] 483. Otter M et al. (2017) [27903454] 533. Rose EM et al. (2009) [19553454]
386. Lee A et al. (2010) [20883814] 434. Metzner L et al. (2005) [16126914] 484. Oude Elferink RP et al. (2007) [16622704] 534. Rosowsky A et al. (2004) [15615544]
387. Lee J et al. (2002) [11734551] 435. Meyer E et al. (2010) [20206334] 485. Owen RP et al. (2006) [16840788] 535. Rotella DP et al. (2009) [19720528]
388. Lee J et al. (2009) [19570976] 436. Mezler M et al. (2008) [18815213] 486. Ozvegy C et al. (2001) [11437380] 536. Rothstein JD et al. (2005) [15635412]
389. Lee JY et al. (2016) [27144356] 437. Michel V et al. (2009) [19357133] 487. Palacín M et al. (1998) [9790568] 537. Rousseau F et al. (2008) [18815261]
390. Lee SG et al. (2008) [18326497] 438. Michel V et al. (2006) [16636297] 488. Pan CJ et al. (2011) [21949678] 538. Ryan RM et al. (2007) [17435767]
391. Lee SH et al. (2008) [18269914] 439. Mihalik SJ et al. (2002) [11980911] 489. Pao SS et al. (1998) [9529885] 539. Ryan RM et al. (2004) [14982939]
392. Lee YC et al. (2010) [20639396] 440. Miki T et al. (1999) [10194514] 490. Paytubi S et al. (2009) [19570978] 540. Rühl A et al. (2005) [16041713]
393. Legati A et al. (2015) [25938945] 441. Milger K et al. (2006) [17062637] 491. Pearlman RJ et al. (2003) [12558979] 541. Sager G et al. (2012) [22380603]
394. Leier I et al. (1994) [7961706] 442. Mimura Y et al. (2017) [28089688] 492. Perland E et al. (2017) [28878041] 542. Sagné C et al. (2001) [11390972]
395. Leung DDM et al. (2010) Anti-ferroportin 443. Mistrík P et al. (2012) [22890707] 493. Perland E et al. (2017) [27939446] 543. Sahni J et al. (2013) [23506895]
monoclonal antibodies and uses thereof. 444. Mitsuoka K et al. (2008) [18344442] 494. Perland E et al. (2016) [27272503] 544. Said HM. (2009) [19056639]
Patent number: WO2010065496 A1. 445. Miura N et al. (1996) [8889805] 495. Pestov NB et al. (2006) [16525125] 545. Said HM et al. (1989) [2911998]
396. Levy LM et al. (1998) [9822723] 446. Miyaji T et al. (2011) [21781115] 496. Pfeffer SR. (2016) [27410046] 546. Saier MH et al. (2009) [19022853]
397. Lewis SE et al. (2001) [11470793] 447. Miyauchi S et al. (2004) [14966140] 497. Pillai SM et al. (2011) [21097500] 547. Saito K et al. (2010) [21190592]
398. Li H et al. (2008) [17928635] 448. Miyazaki E et al. (2001) [11641397] 498. Pinard E et al. (2010) [20491477] 548. Sakata K et al. (2001) [11336635]
399. Li M et al. (2006) [16434549] 449. Mladenova G et al. (2012) [22420844] 499. Pinilla-Tenas J et al. (2003) [14502423] 549. Sala-Rabanal M et al. (2006) [16627568]
400. Li N et al. (2007) [17650074] 450. Morgan RE et al. (2010) [20829430] 500. Pochini L et al. (2014) [24704252] 550. Sala-Rabanal M et al. (2008) [18367661]
401. Lin P et al. (2008) [19061983] 451. Murakami Y et al. (2005) [16174808] 501. Pondarre C et al. (2007) [17192398] 551. Salazar G et al. (2009) [19521526]
402. Lin X et al. (2009) [19032932] 452. Muraoka M et al. (2001) [11322953] 502. Pondarré C et al. (2006) [16467350] 552. Salojin KV et al. (2011) [21346251]
403. Lipovich L et al. (2002) [11943475] 453. Nabulsi NB et al. (2005) [15781409] 503. Potelle S et al. (2017) [28270545] 553. Sandoval A et al. (2010) [19913517]
404. Liu H et al. (2008) [18983139] 454. Nair TS et al. (2004) [14973250] 504. Potelle S et al. (2016) [27008884] 554. Sasawatari S et al. (2011) [21277849]
405. Liu W et al. (1995) [7756356] 455. Nakai Y et al. (2007) [17475902] 505. Prasad PD et al. (1995) [7826387] 555. Sawada K et al. (2008) [18375752]
406. Liu Y et al. (2013) [23597791] 456. Nakamura N et al. (2014) [24695226] 506. Prasad PD et al. (2000) [10772912] 556. Sawada K et al. (1999) [10578127]
407. Liu Z et al. (2008) [18037372] 457. Nakamura N et al. (2005) [15522866] 507. Priebe W et al. (1998) [9647783] 557. Schaffer JE et al. (1994) [7954810]
408. Liuzzi JP et al. (2006) [16950869] 458. Nakamura T et al. (2010) [20410607] 508. Pristupa ZB et al. (1994) [8302271] 558. Schirmer SU et al. (2011) [21482687]
409. Longo N et al. (2016) [26828774] 459. Nakanishi T et al. (2001) [11243884] 509. Pérez-Siles G et al. (2011) [21574997] 559. Schiöth HB et al. (2013) [23506890]
410. Lowe 3rd JA et al. (2003) [12657266] 460. Nawata CM et al. (2015) [26423860] 510. Qiu A et al. (2006) [17129779] 560. Schlipf NA et al. (2010) [20461110]
411. Lowe 3rd JA et al. (2009) [19410451] 461. Neumann J et al. (2003) [12649372] 511. Quamme GA. (2010) [19940067] 561. Schousboe A et al. (2011) [21428813]
412. Lu X et al. (2016) [26494147] 462. Neumann J et al. (2004) [15128310] 512. Quazi F et al. (2014) [24707049] 562. Schousboe A et al. (2004) [15451399]
413. Luckner P et al. (2005) [15567297] 463. Newton JM et al. (2001) [11574907] 513. Quigley JG et al. (2004) [15369674] 563. Secondo A et al. (2015) [25942323]
564. Seidler NW et al. (1989) [2530215] 605. Tamarappoo BK et al. (1996) [8603078] 646. Vavricka SR et al. (2002) [12085361] 687. Yamashita A et al. (2005) [16041361]
565. Sekler I. (2015) [25998733] 606. Tang L et al. (2012) [22085049] 647. Verheijen FW et al. (1999) [10581036] 688. Yamashita K et al. (2016) [27480939]
566. Semyanov A et al. (2004) [15111008] 607. Tanihara Y et al. (2007) [17509534] 648. Veruki ML et al. (2006) [17041592] 689. Yamashita T et al. (1997) [9092568]
567. Sethi AA et al. (2008) [18805791] 608. Tatsumi M et al. (1997) [9537821] 649. Visser WE et al. (2010) [19682536] 690. Yanagisawa H et al. (2003) [12815463]
568. Seyffer F et al. (2015) [24923865] 609. Taylor NMI et al. (2017) [28554189] 650. Voss AA et al. (2007) [17110502] 691. Yao SY et al. (2011) [21795683]
569. Shao J et al. (2016) [27669901] 610. Tejada-Jiménez M et al. (2011) [21464289] 651. Wallgard E et al. (2008) [18483404] 692. Yao Y et al. (2010) [20463145]
570. Shigeri Y et al. (2001) [11677257] 611. Terada T et al. (2006) [16850272] 652. Wang C et al. (2013) [24021350] 693. Yasujima T et al. (2010) [20047987]
571. Shimamoto K et al. (1998) [9463476] 612. Terada T et al. (1997) [9374833] 653. Wang D et al. (2003) [14634667] 694. Ye X et al. (2003) [12670026]
572. Shimamoto K et al. (2007) [17047096] 613. Terada T et al. (1996) [8843163] 654. Wang H et al. (1999) [10329687] 695. Yee BK et al. (2006) [16554468]
573. Shimamoto K et al. (2000) [11078189] 614. Terada T et al. (2000) [10748266] 655. Wang J. (2016) [27506881] 696. Yernool D et al. (2004) [15483603]
574. Shimokawa N et al. (2002) [12417639] 615. Thangaraju M et al. (2006) [16873376] 656. Wang JZ et al. (2016) [26747400] 697. Youngblood GL et al. (2004) [15068970]
575. Shintre CA et al. (2013) [23716676] 616. Thangaraju M et al. (2006) [17178845] 657. Wang Q et al. (2006) [16707723] 698. Yu XC et al. (2009) [19159658]
576. Shu Y et al. (2007) [17476361] 617. Theis S et al. (2002) [11752223] 658. Wang XX et al. (2017) [27845049] 699. Yu Z et al. (2007) [17325024]
577. Sievert MK et al. (1997) [9325342] 618. Thomsen C et al. (1997) [9134205] 659. Wang XX et al. (2018) [29305823] 700. Zaia KA et al. (2009) [19147495]
578. Singer D et al. (2009) [19478081] 619. Thwaites DT et al. (2011) [21501141] 660. Wang Y et al. (2018) [29305856] 701. Zander JF et al. (2010) [20519538]
579. Singh N et al. (2010) [20601425] 620. Tollefson MB et al. (2003) [14552767] 661. Warraich S et al. (2013) [23184610] 702. Zelcer N et al. (2003) [12523936]
580. Singh SK et al. (2007) [17687333] 621. Torres-Salazar D et al. (2007) [17908688] 662. Weinman SA et al. (1998) [9856990] 703. Zeng Z et al. (2008) [18355687]
581. Sloan JL et al. (1999) [10446133] 622. Traiffort E et al. (2005) [15715662] 663. Wenzel U et al. (1998) [9843719]
704. Zerangue N et al. (1996) [8910405]
582. Snyder NA et al. (2019) [30622138] 623. Treiber A et al. (2007) [17496208] 664. Wenzel U et al. (1996) [8627565]
705. Zerangue N et al. (1996) [8857541]
583. Song F et al. (2017) [27836942] 624. Tsai G et al. (2004) [15159536] 665. Westhoff CM et al. (2002) [11861637]
706. Zerangue N et al. (1996) [8782106]
584. Song F et al. (2018) [29224352] 625. Tse CM et al. (1993) [8415663] 666. White C et al. (2013) [23395172]
707. Zhang HX et al. (2009) [19433577]
585. Sreedharan S et al. (2011) [21044875] 626. Tse CM et al. (1993) [7685025] 667. White HS et al. (2005) [15550575]
708. Zhang L et al. (1998) [9655880]
586. Stahl A et al. (1999) [10518211] 627. Tsuda M et al. (2009) [19164462] 668. Wiles AL et al. (2006) [16899062]
709. Zhang X et al. (2009) [19515813]
587. Stansberry WM et al. (2018) [30351207] 628. Tsukaguchi H et al. (1999) [10331392] 669. Wille S et al. (2001) [11698453]
588. Stecula A et al. (2017) [28661652] 629. Tóth A et al. (2002) [12054538] 670. Wilson BJ et al. (2014) [24934811] 710. Zhang Z et al. (2012) [22749870]
589. Stewart G. (2011) [21449978] 630. Uchida Y et al. (2009) [19122366] 671. Wojcik SM et al. (2006) [16701208] 711. Zhao D et al. (2007) [17506977]
590. Stieger B. (2009) [19684528] 631. Uldry M et al. (2002) [12135767] 672. Wolf S et al. (2002) [12049641] 712. Zhao R et al. (2002) [11997266]
591. Sultan M et al. (2018) [28898457] 632. Umapathy NS et al. (2004) [15290873] 673. Wong EH et al. (2000) [10812041] 713. Zheng H et al. (2003) [14756423]
592. Sun D et al. (2013) [23442152] 633. Ussar S et al. (2014) [25080478] 674. Wright EM et al. (2011) [21527736] 714. Zhou H et al. (2009) [19717468]
593. Sundaram M et al. (1998) [9705281] 634. Utsunomiya-Tate N et al. (1996) [8662767] 675. Wright EM et al. (2004) [12748858] 715. Zhou LM et al. (1997) [8996224]
594. Supplisson S et al. (2002) [12354619] 635. van de Wiel SMW et al. (2018) [29675448] 676. Wu CA et al. (2004) [15140889] 716. Zhou M et al. (2010) [20592246]
595. Suzuki H et al. (1998) [9875554] 636. van Leeuwen EM et al. (2015) [25751400] 677. Wu SP et al. (2013) [23259992] 717. Zhou M et al. (2007) [17600084]
596. Suzuki T et al. (2005) [15994300] 637. van Roermund CW et al. (2008) [18757502] 678. Wu X et al. (2002) [12504846] 718. Zhou W et al. (2010) [20448275]
597. Swaan PW et al. (2008) [18474668] 638. van Roermund CW et al. (2011) [21145416] 679. Wu Y et al. (2013) [23678871] 719. Zhu HJ et al. (2010) [20402963]
598. Tagoh H et al. (1996) [8630032] 639. Vandenberg RJ et al. (2004) [15324920] 680. Wängler B et al. (2004) [15380228] 720. Zhu L et al. (2009) [19632829]
599. Tai W et al. (2013) [22950754] 640. Vandenberg RJ et al. (1997) [9145919] 681. Xiang J et al. (2006) [17034769] 721. Zidi-Yahiaoui N et al. (2009) [19553567]
600. Tailor CS et al. (1999) [9927670] 641. Vandenberg RJ et al. (2007) [17383967] 682. Xu X et al. (2018) [29615471] 722. Zimmermann M et al. (2010) [20868728]
601. Tailor CS et al. (1999) [10400745] 642. Vanderperre B et al. (2016) [27317664] 683. Yabuki M et al. (2009) [19236841] 723. Zimmermann M et al. (2010) [19612975]
602. Takeuchi T et al. (2017) [28082679] 643. Vanslambrouck JM et al. (2010) [20377526] 684. Yamada T et al. (2011) [21185344] 724. Zipp GG et al. (2014) [25037917]
603. Talvenheimo J et al. (1983) [6853478] 644. Varoqui H et al. (1996) [8910293] 685. Yamamoto S et al. (2010) [20042597] 725. Zotova L et al. (2010) [20197279]
604. Tamai I et al. (1997) [9379359] 645. Vavricka SR et al. (2004) [15521010] 686. Yamamoto T et al. (1999) [10480349] 726. Zou S et al. (2011) [21426345]