Minireviews Open Access
Copyright ©The Author(s) 2015. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Respirol. Jul 28, 2015; 5(2): 152-159
Published online Jul 28, 2015. doi: 10.5320/wjr.v5.i2.152
Role of hydrogen sulphide in airways
Apostolia Hatziefthimiou, Rodopi Stamatiou, Laboratory of Physiology, Department of Medicine, School of Health Sciences, University of Thessaly, 41110 Larissa, Greece
Author contributions: All authors contributed equally; all authors read and approved the final manuscript for publication.
Conflict-of-interest statement: The authors declare that there is no conflict of interest.
Open-Access: This article is an open-access article which was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution Non Commercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: http://creativecommons.org/licenses/by-nc/4.0/
Correspondence to: Apostolia Hatziefthimiou, Associate Professor of Medical Physiology, Laboratory of Physiology, Department of Medicine, School of Health Sciences, University of Thessaly, Panepistimiou 3, Biopolis, 41110 Larissa, Greece. axatzi@med.uth.gr
Telephone: +30-24-10685561 Fax: +30-24-10685555
Received: December 23, 2014
Peer-review started: December 23, 2014
First decision: January 20, 2015
Revised: February 4, 2015
Accepted: March 5, 2015
Article in press: March 9, 2015
Published online: July 28, 2015

Abstract

The toxicity of hydrogen sulfide (H2S) has been known for a long time, as it is prevalent in the atmosphere. However accumulative data suggest that H2S is also endogenously produced in mammals, including man, and is the third important gas signaling molecule, besides nitric oxide and carbon monoxide. H2S can be produced via non enzymatic pathways, but is mainly synthesized from L-cysteine by the enzymes cystathionine-γ-lyase, cystathionine-β-synthetase, cysteine amino transferase and 3-mercaptopyruvate sulfurtransferase (3MTS). The formation of H2S from D-cysteine via the enzyme D-amino acid oxidase and 3MTS has also been described. Endogenous H2S not only participates in the regulation of physiological functions of the respiratory system, but also seems to contribute to the pathophysiology of airway diseases such as chronic obstructive pulmonary disease, asthma and pulmonary fibrosis, as well as in inflammation, suggesting its possible use as a biomarker for these diseases. This review summarizes the different implications of hydrogen sulfide in the physiology of airways and the pathophysiology of airway diseases.

Key Words: Hydrogen sulfide, Airways, Asthma, Chronic obstructive pulmonary disease, Inflammation

Core tip: Hydrogen sulfide (H2S) is a metabolite produced in mammalian organisms both in physiological and in pathological conditions. The measured levels appear differentiated in inflammatory airway diseases, showing the need to acknowledge H2S not only as a metabolic mediator but as a signaling biomarker as well. This could be of clinical importance since H2S levels could be used in order to access staging or treatment efficiency in patients suffering from airway diseases.



INTRODUCTION

Hydrogen sulfide (H2S) is prevalent in the atmosphere since it is generated from sources both manmade and natural. Therefore organisms may need to either protect themselves from H2S or respond to it, but not particularly in a true signaling way. Although its production in mammalian tissues has been long known, H2S was largely ignored as a metabolic waste. Its toxicity seems to depend mainly on the concentration and considerably less on the duration of H2S exposure[1]. Importantly, accumulating data suggest that H2S is indeed endogenously produced in mammals, including man, and it represents the third important gas-signaling molecule, besides nitric oxide (NO) and carbon monoxide (CO). Even more, a possible interaction between gas-signaling molecules, especially NO and H2S, has been described[2,3]. Endogenously produced H2S affects many biological processes in most human systems, including gastrointestinal, cardiovascular, nervous, endocrine system and kidneys[4-6]. Available data suggest that main cellular targets of H2S are ion channels, such as ATP-sensitive potassium channels (KATP) and transient receptor potential vanilloid channels (TRPV)[7], transcription factors, such as heme oxygenase-1 (HO-1) and nuclear factor kappa B (NF-κB)[8,9], as well as kinases like mitogen-activated protein kinases (MAPK)[10]. These biological effects of H2S have led to the study of its implication in many diseases and the development of H2S-donating drugs with a possible clinical potential[6,11]. The involvement of H2S in the early stages as well as in the development of inflammatory diseases of the respiratory system makes it important to identify it as a biomarker that could be helpful in the prediction or the treatment of such pathological conditions. This review focuses on the effects of H2S on the respiratory system and its implication in airway diseases.

H2S METABOLISM

The metabolic pathways of H2S production in mammals, have been extensively described and are summarized elsewhere[11-13]. Briefly, H2S can be synthesized from L-cysteine, a sulfur-containing amino acid derived from alimentary sources, synthesized from L-methionine through the so-called “trans-sulfuration pathway” with homocysteine as an intermediate, or released from endogenous proteins[1] (Figure 1). H2S is synthesized from L-cysteine by the enzymes cystathionine-γ-lyase (CSE) and cystathionine-β-synthetase (CBS). These enzymes are responsible for the majority of the endogenous production of H2S and their expression appears to be tissue specific[14]. Furthermore, the enzyme cysteine amino transferase (CAT) catalyzes the formation of 3-mercaptopyruvate from L-cysteine that is converted to H2S by the enzyme 3-mercaptopyruvate sulfurtransferase (3MTS). H2S can also be synthesized from D-cysteine via the enzyme D-amino acidoxidase (DAO) that converts D-cysteine to 3-mercaptopyruvate, followed by its conversion to H2S by 3MTS[13]. H2S can also be produced via non enzymatic pathways but these pathways account only for a small portion of the total H2S production[5].

Figure 1
Figure 1 Schematic presentation of pathways for hydrogen sulfide synthesis from L- and D-cysteine via the enzymes cystathionine-γ-lyase, cystathionine-β-synthetase, cysteine amino transferase, D-amino acid oxidase and 3-mercaptopyruvate sulfurtransferase. H2S: Hydrogen sulfide; CSE: Cystathionine-γ-lyase; CBS: Cystathionine-β-synthetase; CAT: Cysteine amino transferase; DAO: D-amino acidoxidase; 3MTS: 3-mercaptopyruvate sulfurtransferase.

H2S, once produced in mammalian cells, can be stored as bound sulfane sulphur and released later in response to a physiological stimulus[12]. H2S is removed quickly from the cellular environment via three main catabolic pathways: (1) H2S oxidation, which takes place mainly in mitochondria, initially to thiosulfate, followed by its conversion to sulfite and sulfate; (2) H2S methylation by thiol S-methyltransferase (TSMT) to methanethiol and dimethylsulfide; and (3) sulfhemoglobin formation by H2S binding to methemoglobin[7] (Figure 2).

Figure 2
Figure 2 Main catabolic pathways of hydrogen sulfide. H2S: Hydrogen sulfide.
H2S EFFECT ON AIRWAYS PHYSIOLOGY

Many different methods have been used in order to estimate H2S physiological levels in the plasma or validate its use as a biomarker for a variety of pathophysiological conditions. This effort is not always easy, due to artifacts, which often lead to inconsistent and contradicting measurements[15]. However, there are studies showing that in healthy adults between the age of 56.6 to 75.0 years, the median H2S serum concentration is approximately 35 μmol/L[16], while H2S plasma concentration seems to be higher in 6-12 years old children (Table 1)[17]. H2S concentration in exhaled air of healthy adult subjects was found to be 8-16 ppb[18]. On the other hand, H2S concentration in lungs, at least in rat, is approximately 30 μmol/L[19]. H2S levels appear altered in some pathological conditions of the airways, like asthma, Chronic Obstructive Pulmonary Disease (COPD) and pneumonia (Table 1) suggesting that H2S is probably involved in the pathophysiology of some airways diseases. Therefore, like exhaled NO, H2S may be a possible biomarker for pulmonary diseases and/or a potential target for new therapeutic approaches for these diseases. Recent studies suggest that H2S participates in the relaxation of airway smooth muscle (Table 2). As far as the ability of airways to produce H2S is concerned, it has only been showed in porcine airways that H2S can be produced endogenously and that the H2S precursor, L-cysteine caused a concentration-dependent relaxation in peripheral bronchioles[20]. Most of the studies concerning the effect of H2S on airways are focused on the effect of exogenous H2S, using H2S donors. Contractility studies regarding the effect of the rapidly releasing H2S donor, sodium hydrosulfide (NaHS) demonstrated that H2S caused a concentration-dependent relaxation in porcine[20], mouse and guinea pig bronchi[21], as well as in rat trachea[22]. This relaxant effect did not depend on epithelium integrity, KATP channels opening or NO release[22]. On the other hand, in guinea pig bronchi[23], as well as mice lung[24], H2S seems to induce the release of sensory neuropeptides, due to the activation of TRPV1 receptors, resulting to the contraction of these bronchi. Therefore, when sensory nerves were desensitized by capsaicin treatment, H2S induced a slight relaxation[23]. Deviations from these findings emerged from the study of Kubo et al[21], which showed that NaHS did not cause contraction in guinea pig bronchi, but a slight relaxation. Recently, an in vivo study revealed that NaHS treatment inhibited the ozone-induced bronchial hyperresponsiveness in mice[25]. Studies regarding the mechanisms involved in H2S-induced relaxation of airways showed that H2S exerts its effect mainly by decreasing intracellular calcium levels. This is due both to reduced calcium influx[26] and to inhibition of Ca2+ release from intracellular stores through InsP3 receptors[27].

Table 1 Hydrogen sulfide concentration in healthy subjects and patients with asthma, chronic obstructive pulmonary disease or pneumonia.
SubjectsAge(yr)H2S concentration in serum/plasma (μmol/L)Ref.
Healthy
71-8035.7 ± 1.2[16]
61-7034.0 ± 0.9[16]
50-6036.4 ± 1.1[16]
64.1 ± 8.735.4 ± 5.3[43]
9.22 ± 1.8052.60 ± 5.56[17]
Patients with bronchial asthma
Bronchial asthma6-1244.17 ± 10.95[17]
Neutrophilic group53.0 ± 13.98.8 ± 4.7[46]
Paucigranulocytic group45.5 ± 15.76.9 ± 2.0[46]
9.03 ± 1.8444.17 ± 10.95[17]
Patients with stable COPD
Patients with acute exacerbations of COPD
73.9 ± 8.333.8 ± 18.6[43]
Patients in stage I to II65.6 ± 1.640.5 ± 6.3[16]
Patients in stage III33.4 ± 2.9[16]
Patients in stage IV27.6 ± 1.6[16]
Patients with pneumonia57.6 ± 20.422.7 ± 14.6[43]
H2S concentration in exhaled air (ppb)
Healthy52.86 ± 19.818.0-16.0[18]
Patients with bronchial asthma
Eosinophilic group46.0 ± 15.27.7 ± 4.2[46]
Paucigranulocytic group45.5 ± 15.711.1 ± 4.6[46]
Patients with COPD
Acute exacerbations67.5 ± 11.478.0-13.0[18]
Stable COPD64.11 ± 8.799.0-12.0[18]
Table 2 The effect of hydrogen sulfide on airway smooth muscle function.
TissueH2S effectsInvolved mechanismRef.
Porcine peripheral bronchiolsRelaxationAlteration in K+ channels activity[20]
Guinea pig main bronchusSlight relaxation[21]
Guinea pig airwaysNeurogenic inflammatory responsesStimulation of TRPV1 receptors on sensory nerves endings[23]
Mouse main bronchusRelaxationIndependent of NK1/NK2 tachykinin receptors, KATP channels, production of NO, cGMP and prostaglandins[21]
Mouse lungNeurogenic inflammationStimulation of NK1 and Substance P release[24]
Mouse small intrapulmonary airwaysRelaxationInhibition of Ca2+ release from intracellular stores through InsP3 receptors[27]
Mouse tracheal smooth muscle cellsRelaxationActivation of BKCa channels[26]
Rat tracheaRelaxationIndependent of KATP channels, β-adrenoceptors, epithelium and production of NO, cGMP and prostaglandins[22]
Human ASMCsRelaxationOpening of KATP channels[29]
Isolated human airway smooth muscle cellsRelaxationInhibition of ERK-1/2 and p38 MAPK phosphorylation[30]
Decrease of cell proliferation and IL-8 release

It has also been shown, that H2S is involved in the relaxation of different smooth muscle types, by affecting a variety of ion channels. For example, in vessels, H2S induces smooth muscle relaxation via its effect on KATP channels located on vascular smooth muscle cells, or on small to medium conductance K+ channels located on vascular endothelial cells, which results to membrane hyperpolarization and smooth muscle relaxation[28]. Similarly, in airways, evidence suggests the implication of K+ channels in the relaxant effect of H2S[20]. Moreover, in primary cultured mouse tracheal smooth muscle cells NaHS seems to activate large conductance calcium activated potassium channels (BKCa) causing an increase in potassium outward currents, cell hyperpolarization and inhibition of Ca2+ influx[26]. Furthermore, H2S caused relaxation by opening KATP channels in isolated human airway smooth muscle cells[29].

Finally, both endogenous and exogenous H2S decreased human airway smooth muscle cell proliferation and interleukin (IL)-8 release induced by FCS, via the inhibition of the phosphorylation of extracellular signal-regulated kinase (ERK)-1/2 and p38 MAPK[30]. The effects of H2S donors that have been described were not affected by the inhibition of CSE, the blockade of KATP channels or NO production.

H2S IN THE PATHOPHYSIOLOGY OF AIRWAY DISEASES

Endogenous H2S participates in the regulation of physiological functions of the respiratory system (Table 3) and seems also to contribute in the pathophysiology of airway diseases such as COPD, asthma and pulmonary fibrosis (Table 3), suggesting its possible use as a biomarker for these diseases. Αpart from the specific features of the pathophysiology, inflammation is a common theme of these diseases. Over the past decade, research data support a key role for H2S in acute or chronic inflammation in different clinical conditions[31] and suggest that H2S has anti-inflammatory and cytoprotective effects that could be beneficial in lung diseases. Animal studies suggest that H2S in the lung increases the anti-inflammatory cytokine, IL-10, while it decreases the pro-inflammatory cytokine, IL-1β in burn and smoke-induced acute lung injury murine models[32] or hyperoxia-induced acute lung injury models in mice[33]. Animal studies also revealed that treatment with H2S attenuated lung injury and prolonged the subjects’ survival[32,33]. Similarly, inhalation of H2S appears to be protective against ventilator-induced lung injury, in mice, by limiting cytokine release and neutrophil transmigration[34]. This protective role is associated with down-regulation of genes related to oxidative stress and inflammation and up-regulation of anti-apoptotic and anti-inflammatory genes[35]. Activating transcription factor 3 (Atf3), a protein that limits pro-inflammatory cytokine expression and controls the balance between proliferative and apoptotic signals[36,37], may have an important role in H2S mediated lung protection, since H2S inhalation up-regulated Atf3 gene[35]. Finally, in mice, NaHS treatment reduced the ozone-induced increase of the total cell number, including neutrophils and macrophages; the levels of cytokines, including tumor necrosis factor-α (TNF-α), chemokine ligand 1, IL-6 and IL-1β[25], as well as the increase of the bronchial alveolar lavage (BAL) fluid. On the other hand, inhalation of H2S protects against ventilator-induced lung injury by preventing edema formation, apoptosis, proinflammatory cytokine production, neutrophil accumulation, and inhibits heme oxygenase-1 expression[34].

Table 3 Implication of hydrogen sulfide in the pathophysiology in human airway diseases - its use as a biomarker.
Disease
COPDHigher serum H2S level in patients with COPD compared with healthy subjects[16]
Acute exacerbation of COPD decreases serum H2S level compared to patients with stable COPD[16,42]
Higher sputum H2S levels in patients with acute exacerbation of COPD compared to those with stable COPD[42]
Higher sputum-to-serum ratio of H2S in COPD subjects with acute exacerbation comparative with those with stable disease[42]
Lower serum H2S levels in patients with COPD who required antibiotics treatment[43]
AsthmaIn children, serum H2S concentration was significantly decreased compared to healthy subjects and correlated positively with FEV1[17]
In adults, exhaled H2S was lowest in eosinophilic asthma correlated positively with FEV1[46]
Pulmonary fibrosisH2S suppress human fibroblast migration, proliferation and phenotype transform stimulated by fetal bovine serum and growth factors and inhibits the TGF-β1-induced differentiation of fibroblasts to myofibroblasts[53]

Although most of the studies suggest that H2S has an anti-inflammatory role, some studies have showed that it may contribute to neurogenic inflammation in airways[38]. Thus, both in guinea pig[23] and mouse[24] H2S induced the release of sensory neuropeptides, while only in mice, it also affected the level of substance P in the lungs, in sepsis-associated lung injury[39].

COPD

Despite inflammation, smoking is the main contributory factor for developing this disease. Animal studies suggest that H2S is protective against smoking-induced lung injury. Namely, exposure of rats to cigarette smoke resulted to an increase in CSE levels and the subsequent H2S administration reduced the number of inflammatory cells, as well as airway hyperresponsiveness[40]. Similar findings were reported in mice with tobacco smoke-induced emphysema[41]. Clinical studies showed evidence that H2S may be implicated in the pathophysiology of COPD and alteration of its levels may be connected with the severity of the disease. In humans, H2S serum levels were significantly higher in patients with COPD compared to healthy subjects and a positive correlation between the severity of COPD and H2S serum levels has been shown. Namely, in patients with stable COPD, H2S serum levels were lower in patients with stage III than in those with stage I obstruction. Additionally, in patients either with or without COPD H2S correlated positively with the percentage of predicted forced expiratory volume (FEV1)[16]. On the other hand, acute exacerbation of COPD decreases H2S serum levels compared to those of patients with stable COPD[16,42]. On the contrary, H2S sputum levels were significantly higher in patients with acute exacerbation of COPD compared to those with stable COPD, which resulted in a higher sputum-to-serum level ratio of H2S in COPD subjects with acute exacerbation in comparison to those with stable disease[42]. As far as COPD treatment is concerned, measured H2S serum levels were significantly lower in patients with COPD who required antibiotics treatment[43], while theophylline treatment did not alter significantly H2S serum levels of COPD patients[44].

Asthma

Clinical studies indicate that H2S serum levels were decreased in patients with either stable asthma or severe acute exacerbations. Even more the changes in H2S serum[45] or exhaled air[25] levels correlated positively with FEV1 and negatively with the count of sputum cells, neutrophils[46] or eosinophils[25]. Similarly in children with asthma, H2S serum concentration was significantly decreased compared to healthy children and the concentration was positively correlated with lung function indices[17]. Whether the decrease of H2S serum levels in patients suffering from asthma is the cause or the consequence is not yet clear. Therefore, it is not clear if H2S levels could be used as a biomarker for the disease, like exhaled NO. However, Tian et al[17] proposed that decreased H2S serum levels might be used to indicate decreasing lung function and Wang et al[45] suggested that nasal H2S could be a way of accurately detecting H2S metabolism in the respiratory system since its levels will not be affected by oral conditions.

Additional evidence for the possible implication of H2S in the pathophysiology of asthma comes from animal studies. In the lungs of OVA-treated rats with asthma H2S serum levels and H2S production from the lungs were decreased in correlation with the decreased CSE expression level and CSE activity in lung tissues[47]. Even more the administration of NaHS or the CSE blocker, D,L-propargylglycine, alleviated or aggravated, respectively, airway hyper-responsiveness in both cigarette smoke exposure model and OVA-induced asthma rat models[40,47].

Pulmonary fibrosis

Pulmonary fibrosis is the final common pathway of a diverse group of lung disorders and is characterized by accumulation and abnormal activation of fibroblasts and myofibroblasts, resulting in excess extracellular matrix deposition and alveolar disruption[48]. Idiopathic pulmonary fibrosis (IPF) is caused by unknown reasons and its pathophysiology has not yet been clarified, while there is a controversy among researchers whether inflammation constitutes the initial stimulus. Nevertheless, the initial stage is quickly followed by abnormal wound healing[49] and the main protein involved in this process seems to be epithelial cell-derived transforming growth factor beta 1 (TGF-β1)[50,51]. Evidence suggests that the endogenous CSE/H2S pathway may participate in the pathogenetic process of pulmonary fibrosis. Myofibroblasts have a main role in the pathogenesis of this disease and although they are generally considered to be differentiated from existing interstitial fibroblasts or bone marrow-derived stem/progenitor cells, epithelial cells also seem to be an important source of myofibroblasts in pulmonary fibrosis[25]. H2S seems to facilitate the maintenance of alveolar epithelial cell phenotype, since TGF-β1 induces epithelial–mesenchymal transition and this effect is suppressed by H2S through a decrease in Smad2/3 phosphorylation, in lungs[52]. Fang et al[53] reported that H2S suppressed human fibroblast migration, proliferation and phenotype transform that was stimulated by fetal bovine serum and growth factors and more specifically inhibited the TGF-β1-induced differentiation to myofibroblasts. These effects on pulmonary fibroblasts were partially mediated by decreased phosphorylation of ERK. Animal studies showed that NaHS administration ameliorated the bleomycin induced pulmonary fibrosis in rats[54,55]. This protective effect of H2S is due, partly, to inhibition of NF-κB p65 expression and regulation of Th1/Th2 balance[55].

Last but not least it is important to point out the potential therapeutic use of H2S. Studies show that both the metabolite itself and its donors could be potentially used in the treatment of various diseases. Specifically, the H2S donor GYY4137 exhibits antihypertensive activity[1], while other donors have anti-inflammatory[56] and antioxidant properties[57]. As far as respiratory diseases are concerned, there are studies showing that the donor-induced elevated levels of H2S are useful in the treatment of respiratory distress syndrome as well as other pathological conditions, since H2S can reduce the oxidative stress that is present in such disorders[1].

CONCLUSION

H2S appears to play a role both in the physiological function and the pathobiological conditions of the respiratory system. Its presence as a metabolite in inflammatory diseases, as well as the correlation that is found between H2S and inflammation mediators such as cytokines or growth factors, support its use as a biomarker of pathological conditions in both the lungs and the airways. However, determining H2S levels in body fluids is not an easy task, because H2S levels are influenced by H2S inhaled from atmospheric air. Such artifacts make its use as a biomarker difficult. Therefore, further studies are required in order to determine the physiological H2S levels and their correlation with the phase, arising or deteriorating, of inflammatory diseases. Overall, it seems that H2S is not a cell waste, but an important metabolite that has yet to receive the proper attention.

ACKNOWLEDGMENTS

We would like to thank Dr. Makris D and Paraskeva E for their critical reading of the manuscript.

Footnotes

P- Reviewer: Abdel-Aziz M, Boggaram V S- Editor: Ji FF L- Editor: A E- Editor: Wang CH

References
1.  Olson KR. The therapeutic potential of hydrogen sulfide: separating hype from hope. Am J Physiol Regul Integr Comp Physiol. 2011;301:R297-R312.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 134]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
2.  Wang R. Two’s company, three’s a crowd: can H2S be the third endogenous gaseous transmitter. FASEB J. 2002;16:1792-1798.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1408]  [Cited by in F6Publishing: 1352]  [Article Influence: 61.5]  [Reference Citation Analysis (0)]
3.  Lo Faro ML, Fox B, Whatmore JL, Winyard PG, Whiteman M. Hydrogen sulfide and nitric oxide interactions in inflammation. Nitric Oxide. 2014;41:38-47.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 138]  [Cited by in F6Publishing: 137]  [Article Influence: 13.7]  [Reference Citation Analysis (0)]
4.  Vandiver M, Snyder SH. Hydrogen sulfide: a gasotransmitter of clinical relevance. J Mol Med (Berl). 2012;90:255-263.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 173]  [Cited by in F6Publishing: 156]  [Article Influence: 13.0]  [Reference Citation Analysis (0)]
5.  Gadalla MM, Snyder SH. Hydrogen sulfide as a gasotransmitter. J Neurochem. 2010;113:14-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 355]  [Cited by in F6Publishing: 336]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
6.  Caliendo G, Cirino G, Santagada V, Wallace JL. Synthesis and biological effects of hydrogen sulfide (H2S): development of H2S-releasing drugs as pharmaceuticals. J Med Chem. 2010;53:6275-6286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 209]  [Article Influence: 14.9]  [Reference Citation Analysis (0)]
7.  Łowicka E, Bełtowski J. Hydrogen sulfide (H2S) - the third gas of interest for pharmacologists. Pharmacol Rep. 2007;59:4-24.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  Oh GS, Pae HO, Lee BS, Kim BN, Kim JM, Kim HR, Jeon SB, Jeon WK, Chae HJ, Chung HT. Hydrogen sulfide inhibits nitric oxide production and nuclear factor-kappaB via heme oxygenase-1 expression in RAW264.7 macrophages stimulated with lipopolysaccharide. Free Radic Biol Med. 2006;41:106-119.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 346]  [Cited by in F6Publishing: 345]  [Article Influence: 19.2]  [Reference Citation Analysis (0)]
9.  Chung HT, Pae HO, Choi BM, Billiar TR, Kim YM. Nitric oxide as a bioregulator of apoptosis. Biochem Biophys Res Commun. 2001;282:1075-1079.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 393]  [Cited by in F6Publishing: 412]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
10.  Du J, Hui Y, Cheung Y, Bin G, Jiang H, Chen X, Tang C. The possible role of hydrogen sulfide as a smooth muscle cell proliferation inhibitor in rat cultured cells. Heart Vessels. 2004;19:75-80.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 139]  [Cited by in F6Publishing: 124]  [Article Influence: 6.2]  [Reference Citation Analysis (0)]
11.  Whiteman M, Le Trionnaire S, Chopra M, Fox B, Whatmore J. Emerging role of hydrogen sulfide in health and disease: critical appraisal of biomarkers and pharmacological tools. Clin Sci (Lond). 2011;121:459-488.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 232]  [Cited by in F6Publishing: 234]  [Article Influence: 18.0]  [Reference Citation Analysis (0)]
12.  Kimura H. The physiological role of hydrogen sulfide and beyond. Nitric Oxide. 2014;41:4-10.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 205]  [Cited by in F6Publishing: 199]  [Article Influence: 19.9]  [Reference Citation Analysis (0)]
13.  Shibuya N, Kimura H. Production of hydrogen sulfide from d-cysteine and its therapeutic potential. Front Endocrinol (Lausanne). 2013;4:87.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 59]  [Cited by in F6Publishing: 66]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
14.  Zhao W, Zhang J, Lu Y, Wang R. The vasorelaxant effect of H(2)S as a novel endogenous gaseous K(ATP) channel opener. EMBO J. 2001;20:6008-6016.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1511]  [Cited by in F6Publishing: 1507]  [Article Influence: 65.5]  [Reference Citation Analysis (0)]
15.  Olson KR, DeLeon ER, Liu F. Controversies and conundrums in hydrogen sulfide biology. Nitric Oxide. 2014;41:11-26.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 103]  [Cited by in F6Publishing: 92]  [Article Influence: 9.2]  [Reference Citation Analysis (0)]
16.  Chen YH, Yao WZ, Geng B, Ding YL, Lu M, Zhao MW, Tang CS. Endogenous hydrogen sulfide in patients with COPD. Chest. 2005;128:3205-3211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 192]  [Cited by in F6Publishing: 185]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
17.  Tian M, Wang Y, Lu YQ, Yan M, Jiang YH, Zhao DY. Correlation between serum H2S and pulmonary function in children with bronchial asthma. Mol Med Rep. 2012;6:335-338.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 35]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
18.  Sun Y, Wang XM, Chen YH, Zhu RX, Liao CC. Exhaled hydrogen sulfide in patients with chronic obstructive pulmonary disease and its correlation with exhaled nitric oxide. Chin Med J (Engl). 2013;126:3240-3244.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 2]  [Reference Citation Analysis (0)]
19.  Dorman DC, Moulin FJ, McManus BE, Mahle KC, James RA, Struve MF. Cytochrome oxidase inhibition induced by acute hydrogen sulfide inhalation: correlation with tissue sulfide concentrations in the rat brain, liver, lung, and nasal epithelium. Toxicol Sci. 2002;65:18-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 289]  [Cited by in F6Publishing: 292]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
20.  Rashid S, Heer JK, Garle MJ, Alexander SP, Roberts RE. Hydrogen sulphide-induced relaxation of porcine peripheral bronchioles. Br J Pharmacol. 2013;168:1902-1910.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 32]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
21.  Kubo S, Doe I, Kurokawa Y, Kawabata A. Hydrogen sulfide causes relaxation in mouse bronchial smooth muscle. J Pharmacol Sci. 2007;104:392-396.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 55]  [Cited by in F6Publishing: 55]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
22.  Gharib-Naseri MK, Saberi S, Mard SA, Latifi SM. Bronchodilatory effect of hydrogen sulfide in rat. Iran J Basic Med Sci. 2012;15:907-915.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Trevisani M, Patacchini R, Nicoletti P, Gatti R, Gazzieri D, Lissi N, Zagli G, Creminon C, Geppetti P, Harrison S. Hydrogen sulfide causes vanilloid receptor 1-mediated neurogenic inflammation in the airways. Br J Pharmacol. 2005;145:1123-1131.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 117]  [Cited by in F6Publishing: 120]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
24.  Bhatia M, Zhi L, Zhang H, Ng SW, Moore PK. Role of substance P in hydrogen sulfide-induced pulmonary inflammation in mice. Am J Physiol Lung Cell Mol Physiol. 2006;291:L896-L904.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 74]  [Cited by in F6Publishing: 80]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
25.  Zhang P, Li F, Wiegman CH, Zhang M, Hong Y, Gong J, Chang Y, Zhang JJ, Adcock I, Chung KF. Inhibitory effect of hydrogen sulfide on ozone-induced airway inflammation, oxidative stress, and bronchial hyperresponsiveness. Am J Respir Cell Mol Biol. 2015;52:129-137.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 32]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
26.  Huang J, Luo YL, Hao Y, Zhang YL, Chen PX, Xu JW, Chen MH, Luo YF, Zhong NS, Xu J. Cellular mechanism underlying hydrogen sulfide induced mouse tracheal smooth muscle relaxation: role of BKCa. Eur J Pharmacol. 2014;741:55-63.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 21]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
27.  Castro-Piedras I, Perez-Zoghbi JF. Hydrogen sulphide inhibits Ca2+ release through InsP3 receptors and relaxes airway smooth muscle. J Physiol. 2013;591:5999-6015.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
28.  Li L, Rose P, Moore PK. Hydrogen sulfide and cell signaling. Annu Rev Pharmacol Toxicol. 2011;51:169-187.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 973]  [Cited by in F6Publishing: 932]  [Article Influence: 71.7]  [Reference Citation Analysis (0)]
29.  Fitzgerald R, DeSantiago B, Lee DY, Yang G, Kim JY, Foster DB, Chan-Li Y, Horton MR, Panettieri RA, Wang R. H2S relaxes isolated human airway smooth muscle cells via the sarcolemmal K(ATP) channel. Biochem Biophys Res Commun. 2014;446:393-398.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 36]  [Cited by in F6Publishing: 29]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
30.  Perry MM, Hui CK, Whiteman M, Wood ME, Adcock I, Kirkham P, Michaeloudes C, Chung KF. Hydrogen sulfide inhibits proliferation and release of IL-8 from human airway smooth muscle cells. Am J Respir Cell Mol Biol. 2011;45:746-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 71]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
31.  Bhatia M. Role of hydrogen sulfide in the pathology of inflammation. Scientifica (Cairo). 2012;2012:159680.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 36]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
32.  Esechie A, Kiss L, Olah G, Horváth EM, Hawkins H, Szabo C, Traber DL. Protective effect of hydrogen sulfide in a murine model of acute lung injury induced by combined burn and smoke inhalation. Clin Sci (Lond). 2008;115:91-97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 93]  [Cited by in F6Publishing: 95]  [Article Influence: 5.9]  [Reference Citation Analysis (0)]
33.  Li HD, Zhang ZR, Zhang QX, Qin ZC, He DM, Chen JS. Treatment with exogenous hydrogen sulfide attenuates hyperoxia-induced acute lung injury in mice. Eur J Appl Physiol. 2013;113:1555-1563.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 33]  [Article Influence: 3.0]  [Reference Citation Analysis (0)]
34.  Faller S, Ryter SW, Choi AM, Loop T, Schmidt R, Hoetzel A. Inhaled hydrogen sulfide protects against ventilator-induced lung injury. Anesthesiology. 2010;113:104-115.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 87]  [Cited by in F6Publishing: 96]  [Article Influence: 6.9]  [Reference Citation Analysis (0)]
35.  Spassov S, Pfeifer D, Strosing K, Ryter S, Hummel M, Faller S, Hoetzel A. Genetic targets of hydrogen sulfide in ventilator-induced lung injury--a microarray study. PLoS One. 2014;9:e102401.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 21]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
36.  Akram A, Han B, Masoom H, Peng C, Lam E, Litvack ML, Bai X, Shan Y, Hai T, Batt J. Activating transcription factor 3 confers protection against ventilator-induced lung injury. Am J Respir Crit Care Med. 2010;182:489-500.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
37.  Thompson MR, Xu D, Williams BR. ATF3 transcription factor and its emerging roles in immunity and cancer. J Mol Med (Berl). 2009;87:1053-1060.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 242]  [Cited by in F6Publishing: 269]  [Article Influence: 17.9]  [Reference Citation Analysis (0)]
38.  Zhang H, Hegde A, Ng SW, Adhikari S, Moochhala SM, Bhatia M. Hydrogen sulfide up-regulates substance P in polymicrobial sepsis-associated lung injury. J Immunol. 2007;179:4153-4160.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 58]  [Cited by in F6Publishing: 63]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
39.  Li L, Bhatia M, Moore PK. Hydrogen sulphide--a novel mediator of inflammation. Curr Opin Pharmacol. 2006;6:125-129.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Chen YH, Wang PP, Wang XM, He YJ, Yao WZ, Qi YF, Tang CS. Involvement of endogenous hydrogen sulfide in cigarette smoke-induced changes in airway responsiveness and inflammation of rat lung. Cytokine. 2011;53:334-341.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 58]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
41.  Han W, Dong Z, Dimitropoulou C, Su Y. Hydrogen sulfide ameliorates tobacco smoke-induced oxidative stress and emphysema in mice. Antioxid Redox Signal. 2011;15:2121-2134.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 92]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
42.  Saito J, Mackay AJ, Rossios C, Gibeon D, Macedo P, Sinharay R, Bhavsar PK, Wedzicha JA, Chung KF. Sputum-to-serum hydrogen sulfide ratio in COPD. Thorax. 2014;69:903-909.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 17]  [Cited by in F6Publishing: 19]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
43.  Chen YH, Yao WZ, Gao JZ, Geng B, Wang PP, Tang CS. Serum hydrogen sulfide as a novel marker predicting bacterial involvement in patients with community-acquired lower respiratory tract infections. Respirology. 2009;14:746-752.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
44.  Chen YH, Yao WZ, Ding YL, Geng B, Lu M, Tang CS. Effect of theophylline on endogenous hydrogen sulfide production in patients with COPD. Pulm Pharmacol Ther. 2008;21:40-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 24]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
45.  Wang P, Zhang G, Wondimu T, Ross B, Wang R. Hydrogen sulfide and asthma. Exp Physiol. 2011;96:847-852.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 66]  [Cited by in F6Publishing: 68]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
46.  Zhang J, Wang X, Chen Y, Yao W. Correlation between levels of exhaled hydrogen sulfide and airway inflammatory phenotype in patients with chronic persistent asthma. Respirology. 2014;19:1165-1169.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 36]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
47.  Chen YH, Wu R, Geng B, Qi YF, Wang PP, Yao WZ, Tang CS. Endogenous hydrogen sulfide reduces airway inflammation and remodeling in a rat model of asthma. Cytokine. 2009;45:117-123.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 122]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
48.  Coward WR, Saini G, Jenkins G. The pathogenesis of idiopathic pulmonary fibrosis. Ther Adv Respir Dis. 2010;4:367-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 203]  [Cited by in F6Publishing: 211]  [Article Influence: 15.1]  [Reference Citation Analysis (0)]
49.  Wilson MS, Wynn TA. Pulmonary fibrosis: pathogenesis, etiology and regulation. Mucosal Immunol. 2009;2:103-121.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 483]  [Cited by in F6Publishing: 549]  [Article Influence: 36.6]  [Reference Citation Analysis (0)]
50.  Khalil N, O’Connor RN, Flanders KC, Unruh H. TGF-beta 1, but not TGF-beta 2 or TGF-beta 3, is differentially present in epithelial cells of advanced pulmonary fibrosis: an immunohistochemical study. Am J Respir Cell Mol Biol. 1996;14:131-138.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 230]  [Cited by in F6Publishing: 233]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
51.  Xu YD, Hua J, Mui A, O’Connor R, Grotendorst G, Khalil N. Release of biologically active TGF-beta1 by alveolar epithelial cells results in pulmonary fibrosis. Am J Physiol Lung Cell Mol Physiol. 2003;285:L527-L539.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 112]  [Cited by in F6Publishing: 125]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
52.  Fang LP, Lin Q, Tang CS, Liu XM. Hydrogen sulfide attenuates epithelial-mesenchymal transition of human alveolar epithelial cells. Pharmacol Res. 2010;61:298-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 31]  [Cited by in F6Publishing: 31]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
53.  Fang LP, Lin Q, Tang CS, Liu XM. Hydrogen sulfide suppresses migration, proliferation and myofibroblast transdifferentiation of human lung fibroblasts. Pulm Pharmacol Ther. 2009;22:554-561.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
54.  Fang L, Li H, Tang C, Geng B, Qi Y, Liu X. Hydrogen sulfide attenuates the pathogenesis of pulmonary fibrosis induced by bleomycin in rats. Can J Physiol Pharmacol. 2009;87:531-538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 39]  [Cited by in F6Publishing: 44]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
55.  Cao H, Zhou X, Zhang J, Huang X, Zhai Y, Zhang X, Chu L. Hydrogen sulfide protects against bleomycin-induced pulmonary fibrosis in rats by inhibiting NF-κB expression and regulating Th1/Th2 balance. Toxicol Lett. 2014;224:387-394.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 46]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
56.  Wallace JL, Dicay M, McKnight W, Martin GR. Hydrogen sulfide enhances ulcer healing in rats. FASEB J. 2007;21:4070-4076.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 172]  [Cited by in F6Publishing: 178]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
57.  Lee M, Tazzari V, Giustarini D, Rossi R, Sparatore A, Del Soldato P, McGeer E, McGeer PL. Effects of hydrogen sulfide-releasing L-DOPA derivatives on glial activation: potential for treating Parkinson disease. J Biol Chem. 2010;285:17318-17328.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 96]  [Cited by in F6Publishing: 86]  [Article Influence: 6.1]  [Reference Citation Analysis (0)]