336
Views
12
CrossRef citations to date
0
Altmetric
Review

Hydrogen sulfide: a target to modulate oxidative stress and neuroplasticity for the treatment of pathological anxiety

ORCID Icon, , , &
Pages 109-121 | Received 27 Feb 2019, Accepted 12 Sep 2019, Published online: 08 Oct 2019

References

  • Kessler RC, Chiu WT, Demler O, et al. Prevalence, severity, and comorbidity of 12-month DSM-IV disorders in the national comorbidity survey replication. Arch Gen Psychiatry. 2005;62:617–627.
  • Hendriks SM, Spijker J, Licht CMM, et al. Long-term disability in anxiety disorders. BMC Psychiatry. 2016;16:248.
  • Judd LL, Kessler RC, Paulus MP, et al. Comorbidity as a fundamental feature of generalized anxiety disorders: results from the national comorbidity study (NCS). Acta Psychiatr Scand Suppl. 1998;393:6–11.
  • Härter MC, Conway KP, Merikangas KR. Associations between anxiety disorders and physical illness. Eur Arch Psychiatry Clin Neurosci. 2003;253:313–320.
  • Lépine J-P. The epidemiology of anxiety disorders: prevalence and societal costs. J Clin Psychiatry. 2002;63(14):4–8.
  • Greenberg PE, Sisitsky T, Kessler RC, et al. The economic burden of anxiety disorders in the 1990s. J Clin Psychiatry. 1999;60:427–435.
  • Liu C, Zhang L, Wu J, et al. AnkG hemizygous mice present cognitive impairment and elevated anxiety/depressive-like traits associated with decreased expression of GABA receptors and postsynaptic density protein. Exp Brain Res. 2017;235:3375–3390.
  • Giuffrè A, Vicente JB. Hydrogen sulfide biochemistry and interplay with other gaseous mediators in mammalian physiology. Oxid Med Cell Longev. 2018;2018:6290931.
  • Gadalla MM, Snyder SH. Hydrogen sulfide as a gasotransmitter. J Neurochem. 2010;113:14–26.
  • Wang R. Hydrogen sulfide: the third gasotransmitter in biology and medicine. Antioxid Redox Signal. 2010;12:1061–1064.
  • Wen Y-D, Wang H, Zhu Y-Z. The Drug Developments of Hydrogen Sulfide on Cardiovascular Disease. Oxid Med Cell Longev. 2018;2018:4010395.
  • Johansen D, Ytrehus K, Baxter GF. Exogenous hydrogen sulfide (H2S) protects against regional myocardial ischemia-reperfusion injury–evidence for a role of K ATP channels. Basic Res Cardiol. 2006;101:53–60.
  • Bełtowski J. Hydrogen sulfide in pharmacology and medicine–an update. Pharmacol Rep. 2015;67:647–658.
  • Paul BD, Snyder SH. Gasotransmitter hydrogen sulfide signaling in neuronal health and disease. Biochem Pharmacol. 2018;149:101–109.
  • Kimura H. Physiological role of hydrogen sulfide and polysulfide in the central nervous system. Neurochem Int. 2013;63:492–497.
  • Reiffenstein RJ, Hulbert WC, Roth SH. Toxicology of hydrogen sulfide. Annu Rev Pharmacol Toxicol. 1992;32:109–134.
  • Olson KR, Straub KD. The role of hydrogen sulfide in evolution and the evolution of hydrogen sulfide in metabolism and signaling. Physiology. 2016;31:60–72.
  • Li Q, Lancaster JR. Chemical foundations of hydrogen sulfide biology. Nitric Oxide Biol Chem. 2013;35:21–34.
  • Paul BD, Snyder SH. H2S: A novel gasotransmitter that signals by sulfhydration. Trends Biochem Sci. 2015;40:687–700.
  • Hosoki R, Matsuki N, Kimura H. The possible role of hydrogen sulfide as an endogenous smooth muscle relaxant in synergy with nitric oxide. Biochem Biophys Res Commun. 1997;237:527–531.
  • Stipanuk MH, Beck PW. Characterization of the enzymic capacity for cysteine desulphhydration in liver and kidney of the rat. Biochem J. 1982;206:267–277.
  • Shibuya N, Tanaka M, Yoshida M, et al. 3-mercaptopyruvate sulfurtransferase produces hydrogen sulfide and bound sulfane sulfur in the brain. Antioxid Redox Signal. 2009;11:703–714.
  • Morikawa T, Kajimura M, Nakamura T, et al. Hypoxic regulation of the cerebral microcirculation is mediated by a carbon monoxide-sensitive hydrogen sulfide pathway. Proc Natl Acad Sci U S A. 2012;109:1293–1298.
  • Mustafa AK, Gadalla MM, Sen N, et al. H2S signals through protein S-sulfhydration. Sci Signal. 2009;2:ra72.
  • Abe K, Kimura H. The possible role of hydrogen sulfide as an endogenous neuromodulator. J Neurosci Off J Soc Neurosci. 1996;16:1066–1071.
  • Whitfield NL, Kreimier EL, Verdial FC, et al. Reappraisal of H2S/sulfide concentration in vertebrate blood and its potential significance in ischemic preconditioning and vascular signaling. Am J Physiol Regul Integr Comp Physiol. 2008;294:R1930–1937.
  • Zhang D, Du J, Tang C, et al. H2S-induced sulfhydration: biological function and detection methodology. Front Pharmacol. 2017;8:608.
  • Prabhakar NR. Sensing hypoxia: physiology, genetics and epigenetics. J Physiol. 2013;591:2245–2257.
  • Guo W, Kan J, Cheng Z, et al. Hydrogen sulfide as an endogenous modulator in mitochondria and mitochondria dysfunction. Oxid Med Cell Longev. 2012;2012:878052.
  • Nagai Y, Tsugane M, Oka J-I, et al. Hydrogen sulfide induces calcium waves in astrocytes. FASEB J Off Publ Fed Am Soc Exp Biol. 2004;18:557–559.
  • Kimura H, Nagai Y, Umemura K, et al. Physiological roles of hydrogen sulfide: synaptic modulation, neuroprotection, and smooth muscle relaxation. Antioxid Redox Signal. 2005;7:795–803.
  • Gutteridge JM. Lipid peroxidation and antioxidants as biomarkers of tissue damage. Clin Chem. 1995;41:1819–1828.
  • Matés JM, Pérez-Gómez C, Núñez de Castro I. Antioxidant enzymes and human diseases. Clin Biochem. 1999;32:595–603.
  • Aladedunye FA, Okorie DA, Ighodaro OM. Anti-inflammatory and antioxidant activities and constituents of platostoma africanum P. Beauv Nat Prod Res. 2008;22:1067–1073.
  • Kerksick C, Willoughby D. The antioxidant role of glutathione and N-acetyl-cysteine supplements and exercise-induced oxidative stress. J Int Soc Sports Nutr. 2005;2:38–44.
  • Meister A. Glutathione deficiency produced by inhibition of its synthesis, and its reversal; applications in research and therapy. Pharmacol Ther. 1991;51:155–194.
  • Zhang J, Wang X, Chen Y, et al. Exhaled hydrogen sulfide predicts airway inflammation phenotype in COPD. Respir Care. 2015;60:251–258.
  • Bazhanov N, Ansar M, Ivanciuc T, et al. Hydrogen sulfide: a novel player in airway development, pathophysiology of respiratory diseases, and antiviral defenses. Am J Respir Cell Mol Biol. 2017;57:403–410.
  • Eto K, Asada T, Arima K, et al. Brain hydrogen sulfide is severely decreased in Alzheimer’s disease. Biochem Biophys Res Commun. 2002;293:1485–1488.
  • Polhemus DJ, Lefer DJ. Emergence of hydrogen sulfide as an endogenous gaseous signaling molecule in cardiovascular disease. Circ Res. 2014;114:730–737.
  • Chan MV, Wallace JL. Hydrogen sulfide-based therapeutics and gastrointestinal diseases: translating physiology to treatments. Am J Physiol Gastrointest Liver Physiol. 2013;305:G467–473.
  • Hovatta I, Tennant RS, Helton R, et al. Glyoxalase 1 and glutathione reductase 1 regulate anxiety in mice. Nature. 2005;438:662–666.
  • Salim S, Sarraj N, Taneja M, et al. Moderate treadmill exercise prevents oxidative stress-induced anxiety-like behavior in rats. Behav Brain Res. 2010;208:545–552.
  • Jang J-H, Surh Y-J. Protective effect of resveratrol on beta-amyloid-induced oxidative PC12 cell death. Free Radic Biol Med. 2003;34:1100–1110.
  • Allam F, Dao AT, Chugh G, et al. Grape powder supplementation prevents oxidative stress-induced anxiety-like behavior, memory impairment, and high blood pressure in rats. J Nutr. 2013;143:835–842.
  • Bouayed J, Rammal H, Younos C, et al. Positive correlation between peripheral blood granulocyte oxidative status and level of anxiety in mice. Eur J Pharmacol. 2007;564:146–149.
  • Rammal H, Bouayed J, Younos C, et al. Evidence that oxidative stress is linked to anxiety-related behaviour in mice. Brain Behav Immun. 2008;22:1156–1159.
  • Nagahara N, Nagano M, Ito T, et al. Antioxidant enzyme, 3-mercaptopyruvate sulfurtransferase-knockout mice exhibit increased anxiety-like behaviors: a model for human mercaptolactate-cysteine disulfiduria. Sci Rep. 2013;3:1986.
  • Kuloglu M, Atmaca M, Tezcan E, et al. Antioxidant enzyme and malondialdehyde levels in patients with panic disorder. Neuropsychobiology. 2002;46:186–189.
  • Ozdemir E, Cetinkaya S, Ersan S, et al. Serum selenium and plasma malondialdehyde levels and antioxidant enzyme activities in patients with obsessive-compulsive disorder. Prog Neuropsychopharmacol Biol Psychiatry. 2009;33:62–65.
  • Kuloglu M, Atmaca M, Tezcan E, et al. Antioxidant enzyme activities and malondialdehyde levels in patients with obsessive-compulsive disorder. Neuropsychobiology. 2002;46:27–32.
  • Kandemir H, Abuhandan M, Aksoy N, et al. Oxidative imbalance in child and adolescent patients with obsessive compulsive disorder. J Psychiatr Res. 2013;47:1831–1834.
  • Atmaca M, Kuloglu M, Tezcan E, et al. Antioxidant enzyme and malondialdehyde levels in patients with social phobia. Psychiatry Res. 2008;159:95–100.
  • Hovatta I, Juhila J, Donner J. Oxidative stress in anxiety and comorbid disorders. Neurosci Res. 2010;68:261–275.
  • Halliwell B. Oxidative stress and neurodegeneration: where are we now? J Neurochem. 2006;97:1634–1658.
  • Koppula P, Zhang Y, Zhuang L, et al. Amino acid transporter SLC7A11/xCT at the crossroads of regulating redox homeostasis and nutrient dependency of cancer. Cancer Commun. 2018;38:12.
  • Jain SK, Huning L, Micinski D. Hydrogen sulfide upregulates glutamate-cysteine ligase catalytic subunit, glutamate-cysteine ligase modifier subunit, and glutathione and inhibits interleukin-1β secretion in monocytes exposed to high glucose levels. Metab Syndr Relat Disord. 2014;12:299–302.
  • Kimura Y, Goto Y-I, Kimura H. Hydrogen sulfide increases glutathione production and suppresses oxidative stress in mitochondria. Antioxid Redox Signal. 2010;12:1–13.
  • Bannai S. Exchange of cystine and glutamate across plasma membrane of human fibroblasts. J Biol Chem. 1986;261:2256–2263.
  • Sun X-L, Zeng X-N, Zhou F, et al. KATP channel openers facilitate glutamate uptake by GluTs in rat primary cultured astrocytes. Neuropsychopharmacol. 2008;33:1336–1342.
  • Lu M, Hu L-F, Hu G, et al. Hydrogen sulfide protects astrocytes against H(2)O(2)-induced neural injury via enhancing glutamate uptake. Free Radic Biol Med. 2008;45:1705–1713.
  • Pan -L-L, Liu X-H, Shen Y-Q, et al. Inhibition of NADPH oxidase 4-related signaling by sodium hydrosulfide attenuates myocardial fibrotic response. Int J Cardiol. 2013;168:3770–3778.
  • Zhang X, Bian J-S. Hydrogen sulfide: a neuromodulator and neuroprotectant in the central nervous system. ACS Chem Neurosci. 2014;5:876–883.
  • Lin MT, Beal MF. Mitochondrial dysfunction and oxidative stress in neurodegenerative diseases. Nature. 2006;443:787–795.
  • Chen W-L, Xie B, Zhang C, et al. Antidepressant-like and anxiolytic-like effects of hydrogen sulfide in behavioral models of depression and anxiety. Behav Pharmacol. 2013;24:590–597.
  • Hu M, Zou W, Wang C-Y, et al. Hydrogen sulfide protects against chronic unpredictable mild stress-induced oxidative stress in hippocampus by upregulation of BDNF-TrkB pathway. Oxid Med Cell Longev. 2016;2016:2153745.
  • Donatti AF, Soriano RN, Leite-Panissi CRA, et al. Anxiolytic-like effect of hydrogen sulfide (H2S) in rats exposed and re-exposed to the elevated plus-maze and open field tests. Neurosci Lett. 2017;642:77–85.
  • Wei H-J, Xu J-H, Li M-H, et al. Hydrogen sulfide inhibits homocysteine-induced endoplasmic reticulum stress and neuronal apoptosis in rat hippocampus via upregulation of the BDNF-TrkB pathway. Acta Pharmacol Sin. 2014;35:707–715.
  • Xu K, Wu F, Xu K, et al. NaHS restores mitochondrial function and inhibits autophagy by activating the PI3K/Akt/mTOR signalling pathway to improve functional recovery after traumatic brain injury. Chem Biol Interact. 2018;286:96–105.
  • Tian Q, Chen L, Luo B, et al. Hydrogen sulfide antagonizes chronic restraint stress-induced depressive-like behaviors via upregulation of adiponectin. Front Psychiatry. 2018;9:399.
  • Kimura H. Hydrogen sulfide induces cyclic AMP and modulates the NMDA receptor. Biochem Biophys Res Commun. 2000;267:129–133.
  • Garrett AM, Schreiner D, Lobas MA, et al. γ-protocadherins control cortical dendrite arborization by regulating the activity of a FAK/PKC/MARCKS signaling pathway. Neuron. 2012;74:269–276.
  • Zheng H, Zhu H-Y, Zhang X-Y, et al. Upregulation of cystathionine β-synthetase in the arcuate nucleus produces pain hypersensitivity via PKC upregulation and GluN2B phosphorylation in rats with chronic pancreatitis. Sheng Li Xue Bao. 2016;68:575–584.
  • Yong QC, Choo CH, Tan BH, et al. Effect of hydrogen sulfide on intracellular calcium homeostasis in neuronal cells. Neurochem Int. 2010;56:508–515.
  • García-Bereguiaín MA, Samhan-Arias AK, Martín-Romero FJ, et al. Hydrogen sulfide raises cytosolic calcium in neurons through activation of L-type Ca2+ channels. Antioxid Redox Signal. 2008;10:31–42.
  • Baker KD, Edwards TM, Rickard NS. The role of intracellular calcium stores in synaptic plasticity and memory consolidation. Neurosci Biobehav Rev. 2013;37:1211–1239.
  • Keeler AB, Schreiner D, Weiner JA. Protein kinase C phosphorylation of a γ-protocadherin C-terminal lipid binding domain regulates focal adhesion kinase inhibition and dendrite arborization. J Biol Chem. 2015;290:20674–20686.
  • Okuno K, Taya K, Marmarou CR, et al. The modulation of aquaporin-4 by using PKC-activator (phorbol myristate acetate) and V1a receptor antagonist (SR49059) following middle cerebral artery occlusion/reperfusion in the rat. Acta Neurochir Suppl. 2008;102:431–436.
  • Wei X, Zhang B, Cheng L, et al. Hydrogen sulfide induces neuroprotection against experimental stroke in rats by down-regulation of AQP4 via activating PKC. Brain Res. 2015;1622:292–299.
  • Saito N, Shirai Y. Protein kinase C gamma (PKC gamma): function of neuron specific isotype. J Biochem. 2002;132:683–687.
  • Miermans CA, Kusters RPT, Hoogenraad CC, et al. Biophysical model of the role of actin remodeling on dendritic spine morphology. PloS One. 2017;12:e0170113.
  • Zhang L-J, Tao -B-B, Wang M-J, et al. PI3K p110α isoform-dependent Rho GTPase Rac1 activation mediates H2S-promoted endothelial cell migration via actin cytoskeleton reorganization. PloS One. 2012;7:e44590.
  • Borovac J, Bosch M, Okamoto K. Regulation of actin dynamics during structural plasticity of dendritic spines: signaling messengers and actin-binding proteins. Mol Cell Neurosci. 2018;91:122–130.
  • Kulkarni VA, Firestein BL. The dendritic tree and brain disorders. Mol Cell Neurosci. 2012;50:10–20.
  • Van Liefferinge J, Bentea E, Demuyser T, et al. Comparative analysis of antibodies to xCT (Slc7a11): forewarned is forearmed. J Comp Neurol. 2016;524:1015–1032.
  • Wang C-M, Yang Y-J, Zhang J-T, et al. Regulation of emotional memory by hydrogen sulfide: role of GluN2B-containing NMDA receptor in the amygdala. J Neurochem. 2015;132:124–134.
  • Chen H-B, Wu W-N, Wang W, et al. Cystathionine-β-synthase-derived hydrogen sulfide is required for amygdalar long-term potentiation and cued fear memory in rats. Pharmacol Biochem Behav. 2017;155:16–23.
  • Shipton OA, Paulsen O. GluN2A and GluN2B subunit-containing NMDA receptors in hippocampal plasticity. Philos Trans R Soc Lond B Biol Sci. 2014;369:20130163.
  • Tovote P, Fadok JP, Lüthi A. Neuronal circuits for fear and anxiety. Nat Rev Neurosci. 2015;16:317–331.
  • Zheng Y, Yu B, De La Cruz LK, et al. Toward hydrogen sulfide based therapeutics: critical drug delivery and developability issues. Med Res Rev. 2018;38:57–100.
  • Whiteman M, Armstrong JS, Chu SH, et al. The novel neuromodulator hydrogen sulfide: an endogenous peroxynitrite “scavenger”? J Neurochem. 2004;90:765–768.
  • Schreier SM, Muellner MK, Steinkellner H, et al. Hydrogen sulfide scavenges the cytotoxic lipid oxidation product 4-HNE. Neurotox Res. 2010;17:249–256.
  • Tay AS, Hu LF, Lu M, et al. Hydrogen sulfide protects neurons against hypoxic injury via stimulation of ATP-sensitive potassium channel/protein kinase C/extracellular signal-regulated kinase/heat shock protein 90 pathway. Neuroscience. 2010;167:277–286.
  • Vannini F, Chattopadhyay M, Kodela R, et al. Positional isomerism markedly affects the growth inhibition of colon cancer cells by NOSH-aspirin: COX inhibition and modeling. Redox Biol. 2015;6:318–325.
  • Lee M, McGeer E, Kodela R, et al. NOSH-aspirin (NBS-1120), a novel nitric oxide and hydrogen sulfide releasing hybrid, attenuates neuroinflammation induced by microglial and astrocytic activation: a new candidate for treatment of neurodegenerative disorders. Glia. 2013;61:1724–1734.
  • Lee M, Tazzari V, Giustarini D, et al. Effects of hydrogen sulfide-releasing L-DOPA derivatives on glial activation: potential for treating Parkinson disease. J Biol Chem. 2010;285:17318–17328.
  • Tang Z-J, Zou W, Yuan J, et al. Antidepressant-like and anxiolytic-like effects of hydrogen sulfide in streptozotocin-induced diabetic rats through inhibition of hippocampal oxidative stress. Behav Pharmacol. 2015;26:427–435.
  • Zhan J-Q, Zheng -L-L, Chen H-B, et al. Hydrogen sulfide reverses aging-associated amygdalar synaptic plasticity and fear memory deficits in rats. Front Neurosci. 2018;12:390.
  • Wallace JL, Wang R. Hydrogen sulfide-based therapeutics: exploiting a unique but ubiquitous gasotransmitter. Nat Rev Drug Discov. 2015;14:329–345.
  • Wallace JL, Caliendo G, Santagada V, et al. Gastrointestinal safety and anti-inflammatory effects of a hydrogen sulfide-releasing diclofenac derivative in the rat. Gastroenterology. 2007;132:261–271.
  • Li L, Rossoni G, Sparatore A, et al. Anti-inflammatory and gastrointestinal effects of a novel diclofenac derivative. Free Radic Biol Med. 2007;42:706–719.
  • Kashfi K, Chattopadhyay M, Kodela R. NOSH-sulindac (AVT-18A) is a novel nitric oxide- and hydrogen sulfide-releasing hybrid that is gastrointestinal safe and has potent anti-inflammatory, analgesic, antipyretic, anti-platelet, and anti-cancer properties. Redox Biol. 2015;6:287–296.
  • Fiorucci S, Orlandi S, Mencarelli A, et al. Enhanced activity of a hydrogen sulphide-releasing derivative of mesalamine (ATB-429) in a mouse model of colitis. Br J Pharmacol. 2007;150:996–1002.
  • Cenac N, Castro M, Desormeaux C, et al. A novel orally administered trimebutine compound (GIC-1001) is anti-nociceptive and features peripheral opioid agonistic activity and Hydrogen Sulphide-releasing capacity in mice. Eur J Pain. 2016;20:723–730.
  • Luoa X, Tong-Yong F, An-lei G, et al. S-adenosyl methionine improves depression-like behaviours and synaptic markers by elevating the expression of endogenous hydrogen sulfide in the hippocampus. Neuropsychiatry. 2018;8:495–504.
  • Zhao Y, Wang H, Xian M. Cysteine-activated hydrogen sulfide (H2S) donors. J Am Chem Soc. 2011;133:15–17.
  • Feng S, Zhao Y, Xian M, et al. Biological thiols-triggered hydrogen sulfide releasing microfibers for tissue engineering applications. Acta Biomater. 2015;27:205–213.
  • Zhao Y, Yang C, Organ C, et al. Design, synthesis, and cardioprotective effects of N-mercapto-based hydrogen sulfide donors. J Med Chem. 2015;58:7501–7511.
  • Foster JC, Radzinski SC, Zou X, et al. H2S-releasing polymer micelles for studying selective cell toxicity. Mol Pharm. 2017;14:1300–1306.
  • Zheng Y, Yu B, Ji K, et al. Esterase-sensitive prodrugs with tunable release rates and direct generation of hydrogen sulfide. Angew Chem Int Ed Engl. 2016;55:4514–4518.
  • Verma R, Akhtar Y, Singh S. A review of patents on therapeutic potential and delivery of hydroge n sulfide. Recent Pat Drug Deliv Formul. 2017;11:114–123.
  • Powell CR, Dillon KM, Matson JB. A review of hydrogen sulfide (H2S) donors: chemistry and potential therapeutic applications. Biochem Pharmacol. 2018;149:110–123.
  • Whiteman M, Li L, Rose P, et al. The effect of hydrogen sulfide donors on lipopolysaccharide-induced formation of inflammatory mediators in macrophages. Antioxid Redox Signal. 2010;12:1147–1154.
  • Amagase H, Petesch BL, Matsuura H, et al. Intake of garlic and its bioactive components. J Nutr. 2001;131:955S–62S.
  • Polhemus DJ, Li Z, Pattillo CB, et al. A novel hydrogen sulfide prodrug, SG1002, promotes hydrogen sulfide and nitric oxide bioavailability in heart failure patients. Cardiovasc Ther. 2015;33:216–226.
  • Shouk R, Abdou A, Shetty K, et al. Mechanisms underlying the antihypertensive effects of garlic bioactives. Nutr Res. 2014;34:106–115.
  • Block E. The chemistry of garlic and onions. Sci Am. 1985;252:114–119.
  • Cai Y-R, Hu C-H. Computational study of H2S release in reactions of diallyl polysulfides with thiols. J Phys Chem B. 2017;121:6359–6366.
  • Tocmo R, Wu Y, Liang D, et al. Boiling enriches the linear polysulfides and the hydrogen sulfide-releasing activity of garlic. Food Chem. 2017;221:1867–1873.
  • Benavides GA, Squadrito GL, Mills RW, et al. Hydrogen sulfide mediates the vasoactivity of garlic. Proc Natl Acad Sci U S A. 2007;104:17977–17982.
  • Rahmani G, Farajdokht F, Mohaddes G, et al. Garlic (Allium sativum) improves anxiety- and depressive-related behaviors and brain oxidative stress in diabetic rats. Arch Physiol Biochem. 2018;125:1–6.
  • Rose P, Moore PK, Zhu Y-Z. Garlic and gaseous mediators. Trends Pharmacol Sci. 2018;39:624–634.
  • DeLeon ER, Gao Y, Huang E, et al. Garlic oil polysulfides: H2S- and O2-independent prooxidants in buffer and antioxidants in cells. Am J Physiol Regul Integr Comp Physiol. 2016;310:R1212–1225.
  • Wang L, Yu H, Zhang Y, et al. Intravenous controlled-release hydrogen sulfide protects against ventilator-induced lung injury. Exp Lung Res. 2017;43:370–377.
  • Louis XL, Murphy R, Thandapilly SJ, et al. Garlic extracts prevent oxidative stress, hypertrophy and apoptosis in cardiomyocytes: a role for nitric oxide and hydrogen sulfide. BMC Complement Altern Med. 2012;12:140.
  • Mostafa DK, El Azhary NM, Nasra RA. The hydrogen sulfide releasing compounds ATB-346 and diallyl trisulfide attenuate streptozotocin-induced cognitive impairment, neuroinflammation, and oxidative stress in rats: involvement of asymmetric dimethylarginine. Can J Physiol Pharmacol. 2016;94:699–708.
  • Ho S-C, Su M-S. Evaluating the anti-neuroinflammatory capacity of raw and steamed garlic as well as five organosulfur compounds. Molecular. 2014;19:17697–17714.
  • You S, Nakanishi E, Kuwata H, et al. Inhibitory effects and molecular mechanisms of garlic organosulfur compounds on the production of inflammatory mediators. Mol Nutr Food Res. 2013;57:2049–2060.
  • Brown RP, Gerbarg PL. Yoga breathing, meditation, and longevity. Ann N Y Acad Sci. 2009;1172:54–62.
  • Last N, Tufts E, Auger LE. The effects of meditation on grey matter atrophy and neurodegeneration: a systematic review. J Alzheimers Dis JAD. 2017;56:275–286.
  • Milad MR, Wright CI, Orr SP, et al. Recall of fear extinction in humans activates the ventromedial prefrontal cortex and hippocampus in concert. Biol Psychiatry. 2007;62:446–454.
  • Hernández SE, Barros-Loscertales A, Xiao Y, et al. Gray matter and functional connectivity in anterior cingulate cortex are associated with the state of mental silence during sahaja yoga meditation. Neuroscience. 2018;371:395–406.
  • Giustino TF, Maren S. The role of the medial prefrontal cortex in the conditioning and extinction of fear. Front Behav Neurosci. 2015;9:298.
  • McCaul KD, Solomon S, Holmes DS. Effects of paced respiration and expectations on physiological and psychological responses to threat. J Pers Soc Psychol. 1979;37:564–571.
  • Djalilova DM, Schulz PS, Berger AM, et al. Impact of Yoga on Inflammatory Biomarkers: A Systematic Review. Biol Res Nurs. 2019;21:198–209.
  • Mohammad A, Thakur P, Kumar R, et al. Biological markers for the effects of yoga as a complementary and alternative medicine. J Complement Integr Med. 2019;16:1.
  • Pan X, Zhang Y, Tao S. Effects of Tai Chi exercise on blood pressure and plasma levels of nitric oxide, carbon monoxide and hydrogen sulfide in real-world patients with essential hypertension. Clin Exp Hypertens. 2015;37:8–14.
  • Meuret AE, Wilhelm FH, Ritz T, et al. Feedback of end-tidal pCO2 as a therapeutic approach for panic disorder. J Psychiatr Res. 2008;42:560–568.
  • Ley R. Panic attacks: klein’s false suffocation alarm, Taylor and Rachman’s data, and Ley’s dyspneic-fear theory. Arch Gen Psychiatry. 1996;53:83–85.
  • Davies CD, McGrath PB, Hale LR, et al. Mediators of change in capnometry guided respiratory intervention for panic disorder. Appl Psychophysiol Biofeedback. 2018;44(2):97–102.
  • McEwen BS. Neurobiological and systemic effects of chronic stress. Chronic Stress. 2017;1:1.
  • Liu S-Y, Li D, Zeng H-Y, et al. hydrogen sulfide inhibits chronic unpredictable mild stress-induced depressive-like behavior by upregulation of Sirt-1: involvement in suppression of hippocampal endoplasmic reticulum stress. Int J Neuropsychopharmacol. 2017;20:867–876.
  • Ercole F, Mansfeld FM, Kavallaris M, et al. Macromolecular hydrogen sulfide donors trigger spatiotemporally confined changes in cell signaling. Biomacromolecules. 2016;17:371–383.
  • Pajares MA, Durán C, Corrales F, et al. Protein kinase C phosphorylation of rat liver S-adenosylmethionine synthetase: dissociation and production of an active monomer. Biochem J. 1994;303(Pt 3):949–955.
  • Mataix-Cols D, Fernández de la Cruz L, Monzani B, et al. D-cycloserine augmentation of exposure-based cognitive behavior therapy for anxiety, obsessive-compulsive, and posttraumatic stress disorders: a systematic review and meta-analysis of individual participant data. JAMA Psychiatry. 2017;74:501–510.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.