The Potential of Hyperbaric Ocygen As A Therapy For Neurodegenerative Diseases
The Potential of Hyperbaric Ocygen As A Therapy For Neurodegenerative Diseases
https://doi.org/10.1007/s11357-022-00707-z
REVIEW
Received: 25 August 2022 / Accepted: 3 December 2022 / Published online: 16 December 2022
© The Author(s), under exclusive licence to American Aging Association 2022
Abstract The World Health Organization estimates Keywords Hyperbaric oxygen · Epigenetics ·
that by the year 2040, neurodegenerative diseases Neurodegenerative diseases · Alzheimer’s disease ·
will be the second leading cause of death in devel- Parkinson’s disease · Oxidative stress · Inflammation ·
oped countries, overtaking cancer-related deaths Neurodegeneration
and exceeded only by cardiovascular disease–related
death. The search for interventions has therefore
become paramount to alleviate some of this burden. Introduction
Based on pathways affected in neurodegenerative dis-
eases, hyperbaric oxygen treatment (HBOT) could Neurodegenerative diseases (NDD) can be charac-
be a good candidate. This therapy has been used for terized by a continuous or progressive loss of a spe-
the past 50 years for conditions such as decompres- cific vulnerable neuronal population in the brain or
sion sickness and wound healing and has been shown spinal cord [1]. The classification of NDD can be
to have promising effects in conditions associated based on the anatomical spread of neurodegenera-
with neurodegeneration and functional impairments. tion (e.g., extrapyramidal disorders, frontotemporal
The goal of this review was to explore the history degenerations, or spinocerebellar degenerations),
of hyperbaric oxygen therapy, its uses, and benefits, the primary molecular abnormality (e.g., amyloid-β,
and to evaluate its effectiveness as an intervention prion protein, tau, α-synuclein), or the main clini-
in treating neurodegenerative diseases. Additionally, cal features (e.g., parkinsonism, motor neuron dis-
we examined common mechanisms underlying the order, or dementia) [1, 2]. Despite these differential
effects of HBOT in different neurodegenerative dis- classifications and symptoms presentations, NDDs,
eases, with a special emphasis on epigenetics. including disorders such as Parkinson’s disease
(PD), amyotrophic lateral sclerosis (ALS), and Alz-
heimer’s disease (AD), share certain processes that
lead to the dysfunction and eventual death of neu-
rons. Some of the major common factors involved
in NDDs are oxidative stress, programmed cell
death, neuroinflammation, mitochondrial dysfunc-
P. Mensah‑Kane · N. Sumien (*) tion, epigenetic modifications, proteotoxic stress,
Department of Pharmacology and Neuroscience, and impairment of its associated ubiquitin/protea-
University of North Texas Health Science Center,
somal and autophagosomal systems [2]. Because of
Fort Worth, TX, USA
e-mail: [email protected]
Vol.: (0123456789)
13
748 GeroScience (2023) 45:747–756
Vol:. (1234567890)
13
GeroScience (2023) 45:747–756 749
Lavoisier and Seguin reported the harmful effects of effects of HBOT are usually dependent on pressure
concentrated oxygen in 1789 [16]. Subsequently, Bert and time exposure [12]. Generally, side effects, such
suggested the use of normobaric instead of hyperbaric as headache, claustrophobia, and reversible myopia,
oxygen for decompression sickness [16]. Behnke and are tolerable and easily reversed when the therapy is
Shaw were the first to utilize HBOT for decompres- halted [21]. Interestingly, HBOT has been used as a
sion sickness treatment in 1937 based on the model tool to induce oxidative stress in screening potential
devised by Drager, who first explored the possibility antioxidants, using high pressure for long periods of
of treating this condition with oxygen under pressure time (above 4 ATA and for a period exceeding 3 h)
[16]. [12, 17].
Vol.: (0123456789)
13
750 GeroScience (2023) 45:747–756
induced by hypoxia involve a complex plethora of magnetic resonance imaging [34]. Also, elderly
factors that tend to be interrelated. They include mito- patients who had significant memory loss exhibited
chondrial dysregulation, oxidative stress, inflamma- improved cognition and increased cerebral blood flow
tion, metal homeostasis, apoptosis, synaptic trans- after exposure to HBOT [35]. In a pre-clinical model
mission, and autophagy, all contributing to neuronal of aging (D-gal-induced aging) in rats [36] and mice
death [22]. Another factor affecting the development [37, 38] as well as in a model of aging and obesity
of dementia and age-related cognitive declines is [36], HBOT prevented cognitive impairments and
the cerebromicrovasculature, as impaired microcir- hippocampal-dependent pathologies via augmenta-
culation has been associated with vascular cognitive tion of cholinergic pathways, anti-apoptotic effects
impairments [30]. Since the fundamental problem in [37], and reduction in oxidative damage and inflam-
the above-described conditions is a dearth of oxygen, matory responses [36, 37].
the theoretical approach to address this problem is to
augment oxygen in the system; therefore, HBOT may Clinical studies of HBOT in neurodegenerative
be a viable tool in their management [30, 31]. diseases
As age is a major risk factor for the development of
several neurodegenerative diseases [32], it is impor- Several clinical studies have been conducted to evalu-
tant to discuss the effect of HBOT on the neurobiol- ate the influence of HBOT on neurodegenerative dis-
ogy of aging. HBOT improved age-related cognitive eases, and of other neurological conditions like stroke
deficits in healthy elderly subjects [33]. In that study, and traumatic brain injury which predisposes patients
male patients with a mean age of 68 years exhibit- to developing neurodegenerative diseases like AD
ing clinical manifestations of intellectual deteriora- (Table 1). In a randomized prospective crossover
tion were assessed after 30 intermittent exposures to control trial of mild traumatic brain injury (mTBI),
100% oxygen at 2.5 ATA. Psychological tests of cog- 56 patients were enrolled and the treated group was
nitive functioning showed highly significant gains in exposed to 40 HBOT sessions (60 min a day at a
treated subjects as compared to control levels [33]. pressure of 1.5 ATA) [39]. The Mindstreams battery
In a recent randomized control clinical trial, healthy test for evaluating cognition, EQ-5D for quality of
adults aged above 64 years were either exposed to life, and SPECT imaging for assessing brain activity
HBOT or no treatment for 3 months. HBOT induced were carried out. Treatment with hyperbaric oxygen
cognitive improvement in these healthy aging adults improved cognitive function and quality of life signif-
through mechanisms involving regional changes in icantly and treated patients also exhibited increased
cerebral blood flow (CBF) measured by perfusion brain activity [39]. In another study, the same research
Table 1 Summary of clinical outcomes of HBOT on brain function and other markers in neurodegenerative diseases
Disease Authors HBOT Findings
Mild traumatic brain injury Boussi-gross et al., 2013 40 sessions at 1.5 ATA, 5 days/ Increased brain activity (SPECT,
week, 60 min/day cognition, and quality of life)
Stroke Boussi-gross et al., 2015 40–60 sessions at 2 ATA, 5 days/ Reduction in memory impairments
week, 90 min/day
Alzheimer’s disease Harch and Fogarty 2018 40 sessions at 1.15 ATA, 5 days/ Increase in energy, brain metabolism
week, 50 min/day and cognition
Alzheimer’s disease Chen et al., 2020 20 sessions at 1.18 ATA, 7 days/ Improved cognitive function
week, 40 min/day (Montreal cognitive assessment,
mini-mental state examination, and
activities of daily living scale)
Parkinson’s disease Xu et al., 2018 30 days at 2 ATA, 2 sessions of Improved non-motor symptoms of
40 min/day separated by 10-min severe depression and anxiety
interval
Amyotrophic lateral sclerosis Steele et al., 2004 20 sessions at 2 ATA, 5 days/week, Decreased fatigue and increased
60 min/day muscle strength
Vol:. (1234567890)
13
GeroScience (2023) 45:747–756 751
group using the same HBOT protocol determined that symptoms of severe depression and anxiety [43].
the treatment led to a reduction in memory impair- Phase I clinical trial involving 5 ALS patients treated
ment in post-stroke patients [40]. In a case study, a with hyperbaric oxygen at 2 atmospheres pressure for
58-year-old woman, diagnosed with rapid progression 60 min daily for 5 days a week for 4 weeks resulted in
AD, was exposed to HBOT for 8 weeks at 1.15 atmos- decreased fatigue and increased muscle strength [44].
phere absolute with 50-min total treatment time, once
per day, 5 days per week [41]. The patient became Pre‑clinical studies of HBOT in neurodegenerative
more energetic and her capacity to carry out tasks like diseases
drawing of clock and daily living was improved after
the 21st day of HBOT. PET imaging showed a corre- There are several pre-clinical studies also suggest-
sponding regional and global increase in brain metab- ing a beneficial role of HBOT in a variety of animal
olism [41]. In a larger clinical study including control models (Table 2). In a mouse model of AD (3 × Tg),
AD patients, AD patients, and amnestic mild cogni- HBOT improved cognition while reducing inflam-
tive impairment (aMCI), psychiatric assessments mation, plaque burden, and Tau phosphorylation [9].
including Montreal cognitive assessment (MoCA), In that study, male 3 × Tg mice aged 17 months were
mini-mental state examination (MMSE), and activi- used with 14-month-old C57BL/6 male mice as con-
ties of daily living (ADL) scale before and at different trols. The treatment with HBO consisted of 2 ATA for
months after treatment were carried out. Treatment an hour daily for 14 consecutive days and resulted in
involved exposing AD and aMCI patients to 40 min diminished neuroinflammatory processes by reducing
of oxygen at a pressure of 0.12 Megapascal (MPa) microgliosis, astrogliosis, and the secretion of pro-
(equivalent of 1.18 ATA) separated by 20-min inter- inflammatory cytokines while improving cognition
vals. HBOT improved cognitive function significantly in the process [9]. In another study, HBOT improved
assessed by MMSE and MoCA in AD patients after cognition in a mouse model of AD (5 × FAD) and
a month follow-up. MMSE score was also improved improved cerebral blood flow [35]. A combination of
by treatment at 3 months while MoCA score saw HBOT and Ginkgo biloba extract has also been found
improvement at both 1- and 3-month follow-ups in to ameliorate cognitive and memory impairment in
aMCI patients [42]. Another case study reported an AD model of rat induced by injecting Aβ 25–35
that HBOT treatment given to Parkinson’s disease [45], through inhibiting oxidative stress by block-
patient over a period of 30 days improved non-motor ing mitochondria-mediated apoptosis signaling [46],
Table 2 Summary of pre-clinical outcomes of HBOT on brain function and other markers in neurodegenerative diseases
Disease Authors HBOT Findings
Alzheimer’s disease-3 × Tg mice Shapira et al., 2018 14 sessions at 2 ATA, 7 days/week, Improved cognition, while reducing
60 min/day inflammation, plaque burden and
Tau phosphorylation
Alzheimer’s disease-5 × FAD Shapira et al., 2021 20 sessions at 2 ATA, 5 days/week, Improved cognition and improved
mice 60 min/day cerebral blood flow
Alzheimer’s disease-Aβ 25–35 Zhang et al., 2015 20 sessions at 2 ATA, (consisted of Improved cognitive and memory
injected rats two courses of 10 days with an impairment in combination with
interval of 3 days between two Ginkgo biloba
courses), 60 min/day
Parkinson’s disease-6-OHDA Pan et al., 2015 14 sessions at 2.4 ATA, 7 days/ Reduced apomorphine-induced turn-
rats week, 60 min/day ing and neuroprotection of substan-
tia nigra’s dopaminergic neurons
Parkinson’s disease-MPTP Hsu et al., 2022 7 sessions at 2.5 ATA, 7 days/ Neuroprotection and improved motor
mouse week, 60 min/day function and increased mitochon-
dria biogenesis signaling
Motor neuron disease-Wobbler Dave et al., 2003 30 days at 2 ATA, 60 min/day Delaying onset via decrease in mito-
mouse chondrial dysfunction
Vol.: (0123456789)
13
752 GeroScience (2023) 45:747–756
activation of NF-κB[45] and enhancing of superoxide both paw condition and walking; from 36 ± 4.3 days
dismutase, an antioxidative enzyme, in rat hippocam- to 59 ± 8.2 days (walking) and from 40 ± 5.7 days to
pal tissue [47]. Pre-treatment of HBOT has also been 63 ± 7.6 days (paw condition) [53].
shown to lower the rate of hippocampal p38 mito-
gen-activated protein kinase phosphorylation with Mechanisms of HBOT
a reduction of hippocampal damage [48]. In a Par-
kinson’s disease rodent model (6-hydroxydopamine HBOT affects diverse cellular and molecular path-
model), the combinatory effect of HBOT and Mado- ways that are important for cellular and neuronal
par (levodopa-used for management of Parkinson’s recovery such as oxidative stress, mitochondrial
disease) therapy was evaluated. Both HBOT and the functions, inflammation, apoptosis, microcircula-
combination therapy reduced apomorphine-induced tion, and epigenetics via interconnected pathway [30,
turning in PD rats and offered neuroprotective effect 31] (Table 3). As aforementioned, these processes
on substantia nigra’s dopaminergic neurons [49]. In and pathways are fundamental and common in most
another model of PD, the 1-methyl-4-phenyl-1,2,3,6- NDDs.
tetrahydropyridine (MPTP) mice, protective mecha-
nisms of HBOT on neurons and motor function were Effect of HBOT on inflammation, oxidative stress,
evaluated. The locomotor activity and grip strength and mitochondrial functions
of the mice were increased alongside increased mito-
chondria biogenesis signaling (SIRT-1, PGC-1α, and HBOT reduces neuroinflammation in severe brain
TFAM) [50]. Furthermore, mild hyperbaric oxygen dysfunctions. HBOT has the capacity to down-
inhibited the decrease of dopaminergic neurons in regulate proinflammatory cytokines (IL-1β, IL-12,
the substantia nigra [51]. The improvement of motor TNFα, and IFNγ) and upregulate an anti-inflamma-
deficits in Parkinson’s disease has also partly been tory cytokine (IL-10) as shown in an atherosclerosis
attributed to the increase in norepinephrine after rodent model [6]. More so, it is reported to inacti-
HBOT exposure [52]. HBOT has also been shown to vate cyclooxygenase, the same target enzyme for
improve function in a model of human motor neuron non-steroidal anti-inflammatory disease [54]. Evi-
disease (Wobbler mouse). HBOT was administered at dence suggests that even though HBOT increases
2 ATA for 1 h daily for 30 days. The treatment signifi- the level of lipid peroxidation and/or protein oxi-
cantly delayed the onset of the disease as measured by dation in blood and tissues of HBOT-exposed
Table 3 Summary of
the mechanisms of action Neuroinflammation Down-regulates pro-inflammatory cytokines (IL-1β,
of hyperbaric oxygen IL-12, TNFα, and IFNγ)
treatment Upregulate an anti-inflammatory cytokine (IL-10, IL-4)
Reduction in astrogliosis and microgliosis
Inactivate cyclooxygenase
Oxidative stress Increase glutathione, superoxide dismutase and catalase
Increase in Nrf2, Heme oxygenase (HO-1)
Reduce cytochrome c release
Neuroprotection and apoptosis Increase mitochondrial transfer from astrocytes to neurons
Increase humanin expression
Increased ATP production and brain metabolism
Increased neurotrophins (BDNF)
Reduction in caspase9/3 mediated apoptotic pathway
Increase in Bcl-2 and decrease in Bax
Microcirculation and neurogenesis Improving of blood–brain barrier permeability
Promotes angiogenesis via VEGF/ERK signaling
Epigenetics Lengthening of telomeres and reduction in cellular senes-
cence
Downregulation of DNA methyltransferase 3a (DNMT3a)
mRNA and protein expression
Vol:. (1234567890)
13
GeroScience (2023) 45:747–756 753
organisms under certain conditions [12, 14], how- a result of the transfer of mitochondria from astro-
ever, HBOT has also been associated with a sig- cytes to neurons [64].
nificant elevation in antioxidants defenses such as
superoxide dismutase, catalase, and glutathione Effect of HBOT on apoptosis and neuroprotection
peroxidase [17]. What is even more fascinating is
that some researchers argue that ROS generated by HBOT has been found to reduce hippocampal p38
HBOT mediates and signals some of the beneficial mitogen-activated protein kinase (MAPK) phospho-
effects of HBOT [14, 17, 55]. Thus, aside from the rylation which led to improvement in cognition and
pressure-dependent effervescence-reducing action reduction in hippocampal damage [48]. Individu-
and the hyperoxygenation saturation of tissues, ally and in combination with Gingko Biloba extract,
the beneficial effect of HBOT has been argued to HBOT reduced Bax expression and the activity of
be in part, dependent on the physiological roles caspase-9/3 leading to a reduction in apoptotic rate in
of the reactive oxygen species generated [56, 57]. Aβ25-35-induced toxicity in rats [46]. In another study,
Superoxide radicals along with hydrogen peroxide HBOT pre-conditioning induced tolerance to cerebral
are known to be the major fraction of ROS pro- ischemia mediated via an increase in SIRT1 with
duced via HBOT [58]. Through a series of steps, an associated increase in B-cell lymphoma 2 Bcl-2)
they activate transcription factors, nuclear factor expression and reduction in cleaved caspase 3 [65].
erythroid 2-related factor 2 (Nrf2), and hypoxia- These corroborate the neuroprotection and anti-apop-
inducible factor-1 alpha (HIF-1α) along with their totic effects of HBOT.
main target protein, heme oxygenase-1 (HO-1), also
called heat shock protein (HSP)32, which provides Effect of HBOT on microcirculation and
robustness to the organism against oxidative dam- neurogenesis
age and plays a vital role in the regulation of cell
proliferation, differentiation, oxidant/antioxidant HBOT exposure has been observed to have a plethora
systems, and apoptosis [59]. HIF-1α is induced in of effects on the cerebrovasculature of the brain [30],
response to hypoxia. Ironically, in HBOT where including improvement of blood–brain barrier perme-
hyperoxia instead of hypoxia is produced, HIF-1α ability [66], angiogenesis [31], and edema reduction
is also induced because of the rapid fall of tissue [30]. HBOT, in addition to providing increased access
oxygen levels after the hyperoxic state, i.e., mimick- of oxygen to tissues, enhances the formation of blood
ing hypoxia. While chronic hypoxia stimulates only vessels via activation of transcription factors such as
HIF-1α, intermittent hypoxia induces both HIF-1α vascular endothelial growth factor as well as neuronal
and Nrf2 [60]. HIF-1α normally downregulates stem cell proliferation [67].
HO-1 induction, but in Nrf2 over-expression, the
inhibitory effect of HIF-1α is reversed, contribut- Effect of HBOT on epigenetics
ing to [the HO-1-mediated action [61]. In accord-
ance, the cycling of hyperoxia-normoxia-hyperoxia It has been found that, after exposure of human
between HBOT sessions can be placed in the con- microvascular endothelial cells (HMEC-1) to HBOT
text of normoxia-hypoxia-normoxia loops, therefore or 24 h later, 8101 genes are significantly regulated
simulating intermittent hypoxia. As a result, HBOT [68]. This includes both up- and downregulation of
stimulates both HIF-1α and Nrf2 [17]. This explains gene expression with increased expression observed
in part the convergence of intermittent hypoxia and in antioxidant response pathways. Hachmo et al. also
HBOT. Mitochondria act as an oxygen sensor by reported that exposure to HBOT led to a lengthening
generating ROS signals through the electron trans- of telomeres and a reduction in cellular senescence
port chain in hypoxic conditions [62]. Following [69]. Shortening of telomeres and increased cellular
HBOT treatment, humanin expression (a neuropro- senescence are two major hallmarks of the aging pro-
tective mitochondrion-derived peptide in humans) cess at the cellular level [69]. What these data suggest
was increased in vascular dementia patients is the capacity of HBOT to exert some influence at
[63]. Recent studies have also suggested that the the genetic level, probably through epigenetic modi-
increased resiliency of neurons exposed to HBOT is fications. Recent evidence suggests dysregulation of
Vol.: (0123456789)
13
754 GeroScience (2023) 45:747–756
epigenetic mechanisms such as DNA methylation, major role. Simply said, more work on the potential
and histone post-translational modifications are impli- of HBOT as a therapy for NDDs needs to be funded
cated in NDDs [70]. Rats exposed to long-term-inter- and completed.
mittent hypoxia exhibited increased DNA methyla-
tion and suppression of several antioxidant enzymes Funding BvB Foundation Dallas; Lowdon Family Founda-
tion; Office of Vice President for Research and Innovation, the
[71]. Also, histone demethylases (chromatin regula- Institute for Healthy Aging, and National Institutes of Health/
tors), such as KDM6A and KDM5A, are oxygen sen- National Institute on Aging (T32 AG020494); GSBS seed
sors that are inactivated by chronic hypoxia [72, 73]. grant; Grant in Aid of Research from the National Academy of
Based on these studies and the fact that the removal Sciences, administered by Sigma Xi, The Scientific Research
Society.
of methyl from lysine and DNA requires oxygen,
we anticipate that one of the major roles of HBOT Declarations
will be through epigenetic modification. In fact, the
pain-relieving effect of HBOT in neuropathic pain is Conflict of interest The authors declare no competing inter-
ests.
believed to be mediated in part through down-regula-
tion of DNA methyltransferase 3a (DNMT3a) mRNA
and protein expression which is an enzyme that plays
a significant role in epigenetic modification [74]. This References
area still remains an unchartered territory and further
1. Kovacs GG. Molecular pathology of neurodegenera-
exploration could hold the key to neurodegenerative
tive diseases: principles and practice. J Clin Pathol.
diseases while answering the questions between the 2019;72(11):725–35.
genetic predisposition of individuals to NDDs and the 2. Dugger BN, Dickson DW. Pathology of neurodegen-
interaction of environmental influences. erative diseases. Cold Spring Harb Perspect Biol.
2017;9(7):a028035.
3. Gammon K. Neurodegenerative disease: brain windfall.
Nature. 2014;515(7526):299–300.
Conclusions 4. Miller E, et al. Potential of redox therapies in neuro-
degenerative disorders. Frontiers in Bioscience-Elite.
2017;9(2):214–34.
The use of HBOT has come a long way. Its availabil-
5. Shah J. Hyperbaric oxygen therapy. J Am Col Certif
ity, reliability, and safety have stood the test of time Wound Spec. 2010;2(1):9–13.
[12, 17]. While the therapy has been used as off label 6. Kudchodkar B, et al. Hyperbaric oxygen treatment
treatment for NDDs and has been somewhat studied, attenuates the pro-inflammatory and immune responses
in apolipoprotein E knockout mice. Clin Immunol.
there is a need for rigorous studies to standardize the
2008;128(3):435–41.
practice of HBOT for NDDs. More studies are needed 7. Huang L, Obenaus A. Hyperbaric oxygen therapy for trau-
to determine how often a patient should receive treat- matic brain injury. Med Gas Res. 2011;1(1):21.
ment, and how long-lasting are the beneficial effects 8. Huang L, et al. Neuroprotective effect of hyperbaric oxy-
gen therapy in a juvenile rat model of repetitive mild trau-
to provide better recommendations on HBO therapy.
matic brain injury. Med Gas Res. 2016;6(4):187–93.
The strength of HBOT as a therapy relies on the mul- 9. Shapira R, et al. Hyperbaric oxygen therapy ameliorates
tiple pathways it seems to affect leading to functional pathophysiology of 3xTg-AD mouse model by attenuating
improvements. The studies of the crucial mechanisms neuroinflammation. Neurobiol Aging. 2018;62:105–19.
10. Hadanny A, Efrati S. The hyperoxic-hypoxic paradox.
that lead to the beneficial effects of HBOT need to
Biomolecules. 2020;10(6):958.
be done to identify therapeutics in case chambers are 11. Waypa GB, Smith KA, Schumacker PT. O2 sensing,
not available, or simply because the treatment itself mitochondria and ROS signaling: the fog is lifting. Mol
is rather inconvenient. Interestingly, there have been Aspects Med. 2016;47:76–89.
12. Simsek K, et al. Evaluation of the oxidative effect of long-
some reported sex differences in using HBOT as an
term repetitive hyperbaric oxygen exposures on different
intervention [75, 76]. This coupled with the preva- brain regions of rats. Sci World J. 2012;2012:849183.
lence or vulnerability of one sex in relation to another 13. Hajhosseini B, et al. Hyperbaric oxygen therapy: descrip-
in neurodegenerative diseases [77, 78] should war- tive review of the technology and current application
in chronic wounds. Plast Reconstr Surg Glob Open.
rant the expansion of future studies to include sex
2020;8(9):e3136.
and possibly race to determine how these factors may 14. Thom SR. Oxidative stress is fundamental to hyperbaric
impact the effect of HBOT and if epigenetics plays a oxygen therapy. J Appl Physiol. 2009;106(3):988–95.
Vol:. (1234567890)
13
GeroScience (2023) 45:747–756 755
15. Jain, K.K., The history of hyperbaric medicine. Text- 36. Shwe T, et al. Hyperbaric oxygen therapy restores
book of hyperbaric medicine, 2017: 3–9 cognitive function and hippocampal pathologies in
16 Edwards ML. Hyperbaric oxygen therapy. Part 1: his- both aging and aging-obese rats. Mech Ageing Dev.
tory and principles. J Vet Emerg Crit Care (San Anto- 2021;195:111465.
nio). 2010;20(3):284–8. 37. Chen C, et al. Hyperbaric oxygen prevents cognitive
17. Simsek K, Sadir S, Oter S. The relation of hyperbaric impairments in mice induced by D-galactose by improv-
oxygen with oxidative stress-reactive molecules in ing cholinergic and anti-apoptotic functions. Neurochem
action. Oxid Antioxid Med Sci. 2015;4(1):17–22. Res. 2017;42(4):1240–53.
18. Cannellotto M, et al. Hyperbaric oxygen as an adjuvant 38. Chen X, et al. Protective effect of hyperbaric oxygen on
treatment for patients with COVID-19 severe hypox- cognitive impairment induced by D-galactose in mice.
aemia: a randomised controlled trial. Emerg Med J. Neurochem Res. 2016;41(11):3032–41.
2022;39(2):88–93. 39. Boussi-Gross R, et al. Hyperbaric oxygen therapy can
19 Levina O, et al. The safety of hyperbaric oxygen therapy improve post concussion syndrome years after mild
in the treatment of COVID-19. Russ Sklifosovsky “J traumatic brain injury-randomized prospective trial.
Emerg Med Care.” 2020;9(3):314–20. PLoS ONE. 2013;8(11):e79995.
20. Leach RM, Rees PJ, Wilmshurst P. Hyperbaric oxygen 40. Boussi-Gross R, et al. Improvement of memory impair-
therapy. BMJ. 1998;317(7166):1140–3. ments in poststroke patients by hyperbaric oxygen ther-
21. Somaa F. A review of the application of hyperbaric oxy- apy. Neuropsychol. 2015;29(4):610–21.
gen therapy in Alzheimer’s Disease. J Alzheimers Dis. 41. Harch PG, Fogarty EF. Hyperbaric oxygen therapy
2021;81(4):1361–7. for Alzheimer’s dementia with positron emission
22. Bhatia D, et al. Hypoxia and its emerging therapeutics tomography imaging: a case report. Med Gas Res.
in neurodegenerative, inflammatory and renal diseases. 2018;8(4):181.
Hypoxia Hum Dis. 2017;21:404–43. 42. Chen J, et al. Hyperbaric oxygen ameliorates cogni-
23. Zhang Z, et al. Hypoxia inducible factor-1 as a tar- tive impairment in patients with Alzheimer’s disease
get for neurodegenerative diseases. Curr Med Chem. and amnestic mild cognitive impairment. Alzheimers
2011;18(28):4335–43. Dement (N Y). 2020;6(1):e12030.
24. Altieri M, et al. Delayed poststroke dementia: a 4-year 43. Xu JJ, et al. Hyperbaric oxygen treatment for Parkin-
follow-up study. Neurol. 2004;62(12):2193–7. son’s disease with severe depression and anxiety: a case
25. Honig LS, Kukull W, Mayeux R. Atherosclerosis and report. Med (Baltimore). 2018;97(9):e0029.
AD: analysis of data from the US National Alzheimer’s 44. Steele J, et al. A Phase I safety study of hyperbaric
Coordinating Center. Neurol. 2005;64(3):494–500. oxygen therapy for amyotrophic lateral sclerosis.
26. Chao LL, et al. ASL perfusion MRI predicts cognitive Amyotroph Lateral Scler Other Motor Neuron Disord.
decline and conversion from MCI to dementia. Alzhei- 2004;5(4):250–4.
mer Dis Assoc Disord. 2010;24(1):19–27. 45. Zhang LD, et al. Hyperbaric oxygen and ginkgo biloba
27. Chen JJ, Rosas HD, Salat DH. Age-associated reduc- extract ameliorate cognitive and memory impairment via
tions in cerebral blood flow are independent from nuclear factor kappa-B pathway in rat model of Alzhei-
regional atrophy. Neuroimage. 2011;55(2):468–78. mer’s disease. Chin Med J (Engl). 2015;128(22):3088–93.
28. Roher AE, et al. Cerebral blood flow in Alzheimer’s dis- 46. Tian X, et al. The protective effect of hyperbaric oxygen
ease. Vasc Health Risk Manag. 2012;8:599–611. and Ginkgo biloba extract on Aβ25–35-induced oxidative
29. Zhang X, Le W. Pathological role of hypoxia in Alzhei- stress and neuronal apoptosis in rats. Behav Brain Res.
mer’s disease. Exp Neurol. 2010;223(2):299–303. 2013;242:1–8.
30. Balasubramanian P, et al. Integrative role of hyper- 47. Tian X, et al. Hyperbaric oxygen and Ginkgo Biloba
baric oxygen therapy on healthspan, age-related vascu- extract inhibit Aβ25-35-induced toxicity and oxidative
lar cognitive impairment, and dementia. Front Aging. stress in vivo: a potential role in Alzheimer’s disease. Int J
2021;2:678543. Neurosci. 2012;122(10):563–9.
31. Gottfried I, Schottlender N, Ashery U. Hyperbaric oxy- 48. Zhao B, et al. Hyperbaric oxygen pretreatment improves
gen treatment-from mechanisms to cognitive improve- cognition and reduces hippocampal damage via p38 mito-
ment. Biomolecules. 2021;11(10):1520. gen-activated protein kinase in a rat model. Yonsei Med J.
32. Hou Y, et al. Ageing as a risk factor for neurodegenera- 2017;58(1):131–8.
tive disease. Nat Rev Neurol. 2019;15(10):565–81. 49. Pan X, et al. Neuroprotective effect of combined therapy
33. Jacobs EA, et al. Hyperoxygenation effect on cog- with hyperbaric oxygen and madopar on 6-hydroxydopa-
nitive functioning in the aged. N Engl J Med. mine-induced Parkinson’s disease in rats. Neurosci Lett.
1969;281(14):753–7. 2015;600:220–5.
34. Hadanny A, et al. Cognitive enhancement of 50. Hsu HT, et al. Hyperbaric oxygen therapy improves Par-
healthy older adults using hyperbaric oxygen: a kinson’s disease by promoting mitochondrial biogen-
randomized controlled trial. Aging (Albany NY). esis via the SIRT-1/PGC-1α pathway. Biomolecules.
2020;12(13):13740–61. 2022;12(5):661.
35. Shapira R, et al. Hyperbaric oxygen therapy alleviates 51. Kusuda Y, et al. Mild hyperbaric oxygen inhibits the
vascular dysfunction and amyloid burden in an Alzhei- decrease of dopaminergic neurons in the substantia nigra
mer’s disease mouse model and in elderly patients. Aging of mice with MPTP-induced Parkinson’s disease. Neuro-
(Albany NY). 2021;13(17):20935–61. sci Res. 2018;132:58–62.
Vol.: (0123456789)
13
756 GeroScience (2023) 45:747–756
52. Banou E. Hyperbaric oxygen therapy effect on “Kine- hypoxic-ischemic brain damage in neonatal rats. Undersea
sia Paradoxa” brain circuits. Adv Exp Med Biol. Hyperb Med. 2008;35(2):113–29.
2021;1339:139–46. 68. Godman CA, et al. Hyperbaric oxygen induces a cyto-
53. Dave K, et al. Hyperbaric oxygen therapy protects protective and angiogenic response in human micro-
against mitochondrial dysfunction and delays onset vascular endothelial cells. Cell Stress Chaperones.
of motor neuron disease in Wobbler mice. Neurosci. 2010;15(4):431–42.
2003;120(1):113–20. 69. Hachmo Y, et al. Hyperbaric oxygen therapy increases
54. Jukic I, et al. Mechanisms of HBO-induced vascu- telomere length and decreases immunosenescence in iso-
lar functional changes in diabetic animal models. In: lated blood cells: a prospective trial. Aging (Albany NY).
Drenjančević I, editor., et al., Hyperbaric Oxygen Treat- 2020;12(22):22445.
ment in Research andClinical Practice. IntechOpen: 70. Hwang J-Y, Aromolaran KA, Zukin RS. The emerging
Rijeka, Croatia; 2018. p. 87–108. field of epigenetics in neurodegeneration and neuroprotec-
55. Thom SR. Hyperbaric oxygen–its mechanisms and effi- tion. Nat Rev Neurosci. 2017;18(6):347–61.
cacy. Plast Reconstr Surg. 2011;127(Suppl 1):131S. 71. Nanduri J, Semenza GL, Prabhakar NR. Epigenetic
56. Dröge W. Free radicals in the physiological control of cell changes by DNA methylation in chronic and intermit-
function. Physiol Rev. 2002;82(1):47–95. tent hypoxia. Am J Physiol-Lung Cell Mol Physiol.
57. Forman HJ, Torres M. Reactive oxygen species and cell 2017;313(6):L1096–100.
signaling: respiratory burst in macrophage signaling. Am 72. Chakraborty AA, et al. Histone demethylase KDM6A
J Respir Crit Care Med. 2002;166(1):S4–8. directly senses oxygen to control chromatin and cell fate.
58. Hink J, Jansen E. Are superoxide and/or hydrogen per- Sci. 2019;363(6432):1217–22.
oxide responsible for some of the beneficial effects 73. Batie M, et al. Hypoxia induces rapid changes to his-
of hyperbaric oxygen therapy? Med Hypotheses. tone methylation and reprograms chromatin. Sci.
2001;57(6):764–9. 2019;363(6432):1222–6.
59. Grochot-Przeczek A, Dulak J, Jozkowicz A. Haem oxyge- 74. Liu K, Wu H, Gao R, Han G. DNA methylation may
nase-1: non-canonical roles in physiology and pathology. be involved in the analgesic effect of hyperbaric oxy-
Clin Sci. 2012;122(3):93–103. gen via regulating FUNDC1. Pain Res Manag.
60. Malec V, et al. HIF-1α signaling is augmented during 2020;2020:1528362. https://doi.org/10.1155/2020/15283
intermittent hypoxia by induction of the Nrf2 pathway in 62
NOX1-expressing adenocarcinoma A549 cells. Free Radi- 75. Brewer A, et al. (308) Investigation of the effect of sex on
cal Biol Med. 2010;48(12):1626–35. hyperbaric oxygen (HBO2)-induced antinociception in a
61. Loboda A, et al. HIF-1 induction attenuates Nrf2-depend- rat model of neuropathic pain. J Pain. 2016;17(4):S53.
ent IL-8 expression in human endothelial cells. Antioxid 76. Huang C-C, et al. Effects of hyperbaric oxygen therapy on
Redox Signal. 2009;11(7):1501–17. acute myocardial infarction following carbon monoxide
62. Hadanny A, Efrati S. The hyperoxic-hypoxic paradox Bio- poisoning. Cardiovasc Toxicol. 2020;20(3):291–300.
molecules. 2020;10(6):958. 77. Pike CJ. Sex and the development of Alzheimer’s disease.
63. Xu Y, et al. Protective effect of hyperbaric oxygen therapy J Neurosci Res. 2017;95(1–2):671–80.
on cognitive function in patients with vascular dementia. 78. Gillies GE, et al. Sex differences in Parkinson’s disease.
Cell Transplant. 2019;28(8):1071–5. Front Neuroendocrinol. 2014;35(3):370–84.
64. Lippert T, Borlongan CV. Prophylactic treatment of
hyperbaric oxygen treatment mitigates inflammatory Publisher’s note Springer Nature remains neutral with regard
response via mitochondria transfer. CNS Neurosci Ther. to jurisdictional claims in published maps and institutional
2019;25(8):815–23. affiliations.
65. Yan W, et al. SirT1 mediates hyperbaric oxygen precon-
ditioning-induced ischemic tolerance in rat brain. J Cereb
Springer Nature or its licensor (e.g. a society or other partner)
Blood Flow Metab. 2013;33(3):396–406.
holds exclusive rights to this article under a publishing
66. Li HZ, et al. Effect of hyperbaric oxygen on the perme-
agreement with the author(s) or other rightsholder(s); author
ability of the blood-brain barrier in rats with global cer-
self-archiving of the accepted manuscript version of this article
ebral ischemia/reperfusion injury. Biomed Pharmacother.
is solely governed by the terms of such publishing agreement
2018;108:1725–30.
and applicable law.
67. Yang YJ, et al. Hyperbaric oxygen induces endog-
enous neural stem cells to proliferate and differentiate in
Vol:. (1234567890)
13