51
|
Working with "H2S": facts and apparent artifacts. Nitric Oxide 2014; 41:85-96. [PMID: 24932545 DOI: 10.1016/j.niox.2014.06.003] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 05/09/2014] [Accepted: 06/06/2014] [Indexed: 01/19/2023]
Abstract
Hydrogen sulfide (H2S) is an important signaling molecule with physiological endpoints similar to those of nitric oxide (NO). Growing interest in its physiological roles and pharmacological potential has led to large sets of contradictory data. The principle cause of these discrepancies can be the common neglect of some of the basic H2S chemistry. This study investigates how the experimental outcome when working with H2S depends on its source and dose and the methodology employed. We show that commercially available NaHS should be avoided and that traces of metal ions should be removed because these can reduce intramolecular disulfides and change protein structure. Furthermore, high H2S concentrations may lead to a complete inhibition of cell respiration, mitochondrial membrane potential depolarization and superoxide generation, which should be considered when discussing the biological effects observed upon treatment with high concentrations of H2S. In addition, we provide chemical evidence that H2S can directly react with superoxide. H2S is also capable of reducing cytochrome c(3+) with the concomitant formation of superoxide. H2S does not directly react with nitrite but with NO electrodes that detect H2S. In addition, H2S interferes with the Griess reaction and should therefore be removed from the solution by Cd(2+) or Zn(2+) precipitation prior to nitrite quantification. 2-Phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide (PTIO) is reduced by H2S, and its use should be avoided in combination with H2S. All these constraints must be taken into account when working with H2S to ensure valid data.
Collapse
|
52
|
Hancock JT, Whiteman M. Hydrogen sulfide and cell signaling: team player or referee? PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2014; 78:37-42. [PMID: 24607577 DOI: 10.1016/j.plaphy.2014.02.012] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/16/2014] [Indexed: 05/09/2023]
Abstract
Hydrogen sulfide (H2S) has been postulated to be the third gasotransmitter, and along with other reactive compounds such as reactive oxygen species (ROS) and nitric oxide (NO) it is thought to be a key signalling molecule. Enzymes which generate H2S, and remove it, have been characterised in both plants and animals and although it is inherently toxic to cells - inhibiting cytochrome oxidase for example - H2S is now being thought of as part of signal transduction pathways. But is it working as a signal in the sense usually seen for small signalling molecules, that is, produced when needed, perceived and leading to dedicated responses in cells? A look through the literature shows that H2S is involved in many stress responses, and in animals is implicated in the onset of many diseases, in both cases where ROS and NO are often involved. It is suggested here that H2S is not acting as a true signal, but through its interaction with NO and ROS metabolism is modulating such activity, keeping it in check unless strictly needed, and that H2S is acting as a referee to ensure NO and ROS metabolism is working properly.
Collapse
Affiliation(s)
- J T Hancock
- Faculty of Health and Applied Sciences, University of the West of England, Coldharbour Lane, Bristol BS16 1QY, UK.
| | - M Whiteman
- University of Exeter Medical School, University of Exeter, Exeter, UK
| |
Collapse
|
53
|
Moustafa A, Habara Y. Hydrogen sulfide regulates Ca(2+) homeostasis mediated by concomitantly produced nitric oxide via a novel synergistic pathway in exocrine pancreas. Antioxid Redox Signal 2014; 20:747-58. [PMID: 24138560 PMCID: PMC3910447 DOI: 10.1089/ars.2012.5108] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
AIM The present study was designed to explore the effects of hydrogen sulfide (H2S) on Ca(2+) homeostasis in rat pancreatic acini. RESULTS Sodium hydrosulfide (NaHS; an H2S donor) induced a biphasic increase in the intracellular Ca(2+) concentration ([Ca(2+)]i) in a dose-dependent manner. The NaHS-induced [Ca(2+)]i elevation persisted with an EC50 of 73.3 μM in the absence of extracellular Ca(2+) but was abolished by thapsigargin, indicating that both Ca(2+) entry and Ca(2+) release contributed to the increase. The [Ca(2+)]i increase was markedly inhibited in the presence of NG-monomethyl L-arginine or 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (cPTIO), and diaminofluorescein-2/diaminofluorescein-2 triazole (DAF-2/DAF-2T) fluorometry demonstrated that nitric oxide (NO) was also produced by H2S in a dose-dependent manner with an EC50 of 64.8 μM, indicating that NO was involved in the H2S effect. The H2S-induced [Ca(2+)]i increase was inhibited by pretreatment with U73122, xestospongin C, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, KT5823, and GP2A, indicating that phospholipase C (PLC), the inositol 1,4,5-trisphosphate (IP3) receptor, soluble guanylate cyclase (sGC), protein kinase G (PKG), and Gq-protein play roles as intermediate components in the H2S-triggered intracellular signaling. INNOVATION To our knowledge, our study is the first one highlighting the effect of H2S on intracellular Ca(2+) dynamics in pancreatic acinar cells. Moreover, a novel cascade was presumed to function via the synergistic interaction between H2S and NO. CONCLUSION We conclude that H2S affects [Ca(2+)]i homeostasis that is mediated by H2S-evoked NO production via an endothelial nitric oxide synthase (eNOS)-NO-sGC-cyclic guanosine monophosphate-PKG-Gq-protein-PLC-IP3 pathway to induce Ca(2+) release, and this pathway is identical to the one we recently proposed for a sole effect of NO and the two gaseous molecules synergistically function to regulate Ca(2+) homeostasis.
Collapse
Affiliation(s)
- Amira Moustafa
- 1 Laboratory of Physiology, Department of Biomedical Sciences, Graduate School of Veterinary Medicine, Hokkaido University , Sapporo, Japan
| | | |
Collapse
|
54
|
Sha L, Linden DR, Farrugia G, Szurszewski JH. Effect of endogenous hydrogen sulfide on the transwall gradient of the mouse colon circular smooth muscle. J Physiol 2013; 592:1077-89. [PMID: 24366262 DOI: 10.1113/jphysiol.2013.266841] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A transwall gradient in resting membrane potential (RMP) exists across the circular muscle layer in the mouse colon. This gradient is dependent on endogenous generation of CO. H2S is also generated in muscle layers of the mouse colon. The effect of endogenously generated H2S on the transwall gradient is not known. The aim was to investigate the role of endogenous H2S. Our results showed that the CSE inhibitor dl-propargylglycine (PAG, 500 μm) had no effect on the transwall gradient. However, in preparations pretreated with the nitric oxide synthase inhibitor N-nitro-l-arginine (l-NNA, 200 μm) and in nNOS-knockout (KO) mouse preparations, PAG shifted the transwall gradient in the depolarizing direction. In CSE-KO-nNOS-KO mice, the gradient was shifted in the depolarizing direction. Endogenous generation of NO was significantly higher in muscle preparations of CSE-KO mice compared to wild-type (WT) mice. The amplitude of NO-mediated slow inhibitory junction potentials (S-IJPs) evoked by electric field stimulation was significantly higher in CSE-KO mouse preparations compared to the amplitude of S-IJPs in wild-type mouse preparations. CSE was present in all submucosal ganglion neurons and in almost all myenteric ganglion neurons. Eleven per cent of CSE positive neurons in the submucosal plexus and 50% of CSE positive neurons in the myenteric plexus also contained nNOS. Our results suggest that endogenously generated H2S acts as a stealth hyperpolarizing factor on smooth muscle cells to maintain the CO-dependent transwall gradient and inhibits NO production from nNOS.
Collapse
Affiliation(s)
- L Sha
- Department of Physiology and Biomedical Engineering, Mayo Clinic, 200 First Street SW, Rochester, MN 55905 USA. E-mail:
| | | | | | | |
Collapse
|
55
|
Abstract
Hydrogen sulfide (H2S), a gas characterized by the odor of rotten eggs, is produced by many cells in the airways and lungs, and may regulate physiologic and pathophysiologic processes. It plays a role in cellular signaling, and represents the third gasotransmitter after nitric oxide and carbon monoxide. Endogenous and exogenous H₂S have anti-inflammatory and anti-proliferative effects, with inhibitory effects in models of lung inflammation and fibrosis. Under certain conditions, H₂S may also be proinflammatory. It is generally a vasodilator and relaxant of airway and vascular smooth muscle cells. It acts as a reducing agent, being able to scavenge superoxide and peroxynitrite. H₂S is detectable in serum and in sputum supernatants with raised levels observed in asthmatics. The sputum levels correlated inversely with lung function. H₂S may play a role in the pathogenesis of asthma.
Collapse
Affiliation(s)
- Kian F Chung
- National Heart & Lung Institute, Imperial College & NIHR Respiratory Biomedical Research Unit at the Royal Brompton and Harefield NHS Foundation Trust and Imperial College London, UK +44 207 352 8121
| |
Collapse
|
56
|
Lisjak M, Teklic T, Wilson ID, Whiteman M, Hancock JT. Hydrogen sulfide: environmental factor or signalling molecule? PLANT, CELL & ENVIRONMENT 2013; 36:1607-16. [PMID: 23347018 DOI: 10.1111/pce.12073] [Citation(s) in RCA: 150] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2012] [Revised: 01/15/2013] [Accepted: 01/16/2013] [Indexed: 05/19/2023]
Abstract
Hydrogen sulfide (H₂S) has traditionally been thought of as a phytotoxin, having deleterious effects on the plant growth and survival. It is now recognized that plants have enzymes which generate H₂S, cysteine desulfhydrase, and remove it, O-acetylserine lyase. Therefore, it has been suggested that H₂S is considered as a signalling molecule, alongside small reactive compounds such as hydrogen peroxide (H₂O₂) and nitric oxide (NO). Exposure of plants to low of H₂S, for example from H₂S donors, is revealing that many physiological effects are seen. H₂S seems to have effects on stomatal apertures. Intracellular effects include increases in glutathione levels, alterations of enzyme activities and influences on NO and H₂O₂ metabolism. Work in animals has shown that H₂S may have direct effects on thiol modifications of cysteine groups, work that will no doubt inform future studies in plants. It appears therefore, that instead of thinking of H₂S as a phytotoxin, it needs to be considered as a signalling molecule that interacts with reactive oxygen species and NO metabolism, as well as having direct effects on the activity of proteins. The future may see H₂S being used to modulate plant physiology in the field or to protect crops from postharvest spoilage.
Collapse
Affiliation(s)
- Miroslav Lisjak
- Department of Agroecology, University of J. J. Strossmayer, Osijek 31000, Croatia
| | | | | | | | | |
Collapse
|
57
|
Paulsen C, Carroll KS. Cysteine-mediated redox signaling: chemistry, biology, and tools for discovery. Chem Rev 2013; 113:4633-79. [PMID: 23514336 PMCID: PMC4303468 DOI: 10.1021/cr300163e] [Citation(s) in RCA: 868] [Impact Index Per Article: 72.3] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2012] [Indexed: 02/06/2023]
Affiliation(s)
- Candice
E. Paulsen
- Department of Chemistry, The Scripps Research
Institute, Jupiter, Florida, 33458, United States
| | - Kate S. Carroll
- Department of Chemistry, The Scripps Research
Institute, Jupiter, Florida, 33458, United States
| |
Collapse
|
58
|
Kolluru GK, Shen X, Bir SC, Kevil CG. Hydrogen sulfide chemical biology: pathophysiological roles and detection. Nitric Oxide 2013; 35:5-20. [PMID: 23850632 DOI: 10.1016/j.niox.2013.07.002] [Citation(s) in RCA: 328] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2012] [Revised: 06/26/2013] [Accepted: 07/02/2013] [Indexed: 01/02/2023]
Abstract
Hydrogen sulfide (H2S) is the most recent endogenous gasotransmitter that has been reported to serve many physiological and pathological functions in different tissues. Studies over the past decade have revealed that H2S can be synthesized through numerous pathways and its bioavailability regulated through its conversion into different biochemical forms. H2S exerts its biological effects in various manners including redox regulation of protein and small molecular weight thiols, polysulfides, thiosulfate/sulfite, iron-sulfur cluster proteins, and anti-oxidant properties that affect multiple cellular and molecular responses. However, precise measurement of H2S bioavailability and its associated biochemical and pathophysiological roles remains less well understood. In this review, we discuss recent understanding of H2S chemical biology, its relationship to tissue pathophysiological responses and possible therapeutic uses.
Collapse
Affiliation(s)
- Gopi K Kolluru
- Department of Pathology, LSU Health-Shreveport, United States
| | | | | | | |
Collapse
|
59
|
Rubini A, Porzionato A, Zara S, Cataldi A, Garetto G, Bosco G. The effect of acute exposure to hyperbaric oxygen on respiratory system mechanics in the rat. Lung 2013; 191:459-66. [PMID: 23828552 DOI: 10.1007/s00408-013-9488-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 06/14/2013] [Indexed: 02/07/2023]
Abstract
PURPOSE This study was designed to investigate the possible effects of acute hyperbaric hyperoxia on respiratory mechanics of anaesthetised, positive-pressure ventilated rats. METHODS We measured respiratory mechanics by the end-inflation occlusion method in nine rats previously acutely exposed to hyperbaric hyperoxia in a standard fashion. The method allows the measurements of respiratory system elastance and of both the "ohmic" and of the viscoelastic components of airway resistance, which respectively depend on the newtonian pressure dissipation due to the ohmic airway resistance to air flow, and on the viscoelastic pressure dissipation caused by respiratory system tissues stress-relaxation. The activities of inducible and endothelial NO-synthase in the lung's tissues (iNOS and eNOS respectively) also were investigated. Data were compared with those obtained in control animals. RESULTS We found that the exposure to hyperbaric hyperoxia increased respiratory system elastance and both the "ohmic" and viscoelastic components of inspiratory resistances. These changes were accompanied by increased iNOS but not eNOS activities. CONCLUSIONS Hyperbaric hyperoxia was shown to acutely induce detrimental effects on respiratory mechanics. A possible causative role was suggested for increased nitrogen reactive species production because of increased iNOS activity.
Collapse
Affiliation(s)
- Alessandro Rubini
- Section Physiology, Department of Biomedical Sciences, University of Padova, Via Marzolo, 3, 35100, Padua, Italy,
| | | | | | | | | | | |
Collapse
|
60
|
Norris EJ, Feilen N, Nguyen NH, Culberson CR, Shin MC, Fish M, Clemens MG. Hydrogen sulfide modulates sinusoidal constriction and contributes to hepatic microcirculatory dysfunction during endotoxemia. Am J Physiol Gastrointest Liver Physiol 2013; 304:G1070-8. [PMID: 23518685 DOI: 10.1152/ajpgi.00395.2012] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hydrogen sulfide (H₂S) affects vascular resistance; however, its effect on the hepatic microcirculation has not been investigated. Hepatic sinusoidal perfusion is dysregulated during sepsis, contributing to liver injury. Therefore, the present study determined the effect of H₂S on the hepatic microcirculation and the contribution of endogenous H₂S to hepatic microcirculatory dysfunction in an endotoxin model of sepsis. Portal infusion of H₂S increased portal pressure in vivo (6.8 ± 0.2 mmHg before H₂S vs. 8.6 ± 0.8 mmHg peak during H₂S infusion, P < 0.05). Using intravital microscopy, we observed decreased sinusoidal diameter (6.2 ± 0.27 μm before H₂S vs. 5.7 ± 0.3 μm after H₂S, P < 0.05) and increased sinusoidal heterogeneity during H₂S infusion (P < 0.05) and net constriction. Since hepatic H₂S levels are elevated during sepsis, we used the cystathionine γ lyase inhibitor DL-propargylglycine (PAG) to determine the contribution of H₂S to the hypersensitization of the sinusoid to the vasoconstrictor effect of endothelin-1 (ET-1). PAG treatment significantly attenuated the sinusoidal sensitization to ET-1 in endotoxin-treated animals. ET-1 infusion increased portal pressure to 175% of baseline in endotoxemic animals, which was reduced to 143% following PAG treatment (P < 0.05). PAG abrogated the increase in sinusoidal constriction after ET-1 infusion in LPS-treated rats (30.9% reduction in LPS rats vs. 11.6% in PAG/LPS rats, P < 0.05). Moreover, PAG treatment significantly attenuated the increase in NADH fluorescence following ET-1 exposure during endotoxemia (61 grayscale units LPS vs. 21 units in PAG/LPS, P < 0.05), suggesting an improvement in hepatic oxygen availability. This study is the first to demonstrate a vasoconstrictor action of H₂S on the hepatic sinusoid and provides a possible mechanism for the protective effect of PAG treatment during sepsis.
Collapse
Affiliation(s)
- Eric J Norris
- Department of Biology, University of North Carolina at Charlotte, Charlotte, NC 28223, USA
| | | | | | | | | | | | | |
Collapse
|
61
|
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) have emerged as dominant redox regulators of numerous aspects of cellular and physiological functions within several organ systems included cardiovascular, immune and neurological tissues. Recent studies have begun to reveal that these two gaseous molecules may have redundant or overlapping pathophysiological functions often involving similar molecular targets. However, it remains less clear when and how NO and H2S may interact under biological and disease processes. In this graphical review, we discuss the current understanding of NO and H2S interactions and how they may functionally influence each other and what this may mean for biology and mechanisms of disease. H2S and NO are important gaseous regulators of numerous physiological responses. H2S and NO may target both similar and divergent signaling and molecular pathways. H2S and NO react with protein free thiols that differentially affect protein function. H2S and NO metabolites can react together to yield novel biochemical products. The presence and physiological importance of these novel products remains unknown.
Collapse
|
62
|
Ning J, Mo L, Zhao H, Lu K, Lai X, Luo X, Zhao H, Ma D. Sodium Hydrosulphide alleviates remote lung injury following limb traumatic injury in rats. PLoS One 2013; 8:e59100. [PMID: 23527096 PMCID: PMC3602436 DOI: 10.1371/journal.pone.0059100] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2012] [Accepted: 02/11/2013] [Indexed: 12/28/2022] Open
Abstract
Hydrogen sulphide (H2S) was found to attenuate ventilator or oleic acid induced lung injury. The aim of this study was to explore the effects of exogenous H2S donor, sodium Hydrosulphide (NaHS), on lung injury following blast limb trauma and the underlying mechanisms. For in vitro experiments, pulmonary micro-vessel endothelial cells (PMVECs) were cultured and treated with NaHS or vehicle in the presence of TNF-α. For in vivo, blast limb traumatic rats, induced by using chartaceous electricity detonators, were randomly treated with NaHS, cystathionine gamma-lyase inhibitor (PAG) or vehicle. In vitro, NaHS (100 µM) treatment increased PMVECs viability and decreased LDH release into culture media after tumor necrosis factor (TNF) α challenge. In addition, NaHS treatment prevented the increase of nitric oxide, Intercellular Adhesion Molecule 1(ICAM-1) and interleukin (IL)-6 production and inducible nitric oxide synthase activation induced by TNF-α. Knock-down of NF-E2-Related Factor 2 (Nrf2) partially abolished the protective effect of NaHS. In vivo, NaHS treatment significantly alleviated lung injury following blast limb trauma, demonstrated by a decreased histopathological score and lung water content. Furthermore, NaHS treatment reversed the decrease of H2S concentration in plasma, prevented the increase of TNF-α, IL-6, malondialdehyde and myeloperoxidase, increased the Nrf2 downstream effector glutathione in both plasma and lungs, and reversed the decrease of superoxide dismutase in both plasma and lungs induced by blast limb trauma. Our data indicated that NaHS protects against lung injury following blast limb trauma which is likely associated with suppression of the inflammatory and oxidative response and activation of Nrf2 cellular signal.
Collapse
Affiliation(s)
- Jiaolin Ning
- Department of Anesthesiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | |
Collapse
|
63
|
Li L, Fox B, Keeble J, Salto-Tellez M, Winyard PG, Wood ME, Moore PK, Whiteman M. The complex effects of the slow-releasing hydrogen sulfide donor GYY4137 in a model of acute joint inflammation and in human cartilage cells. J Cell Mol Med 2013; 17:365-76. [PMID: 23356870 PMCID: PMC3823018 DOI: 10.1111/jcmm.12016] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2012] [Accepted: 12/04/2012] [Indexed: 01/01/2023] Open
Abstract
The role of hydrogen sulfide (H2S) in inflammation remains unclear with both pro- and anti-inflammatory actions of this gas described. We have now assessed the effect of GYY4137 (a slow-releasing H2S donor) on lipopolysaccharide (LPS)-evoked release of inflammatory mediators from human synoviocytes (HFLS) and articular chondrocytes (HAC) in vitro. We have also examined the effect of GYY4137 in a complete Freund's adjuvant (CFA) model of acute joint inflammation in the mouse. GYY4137 (0.1–0.5 mM) decreased LPS-induced production of nitrite (NO2−), PGE2, TNF-α and IL-6 from HFLS and HAC, reduced the levels and catalytic activity of inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) and reduced LPS-induced NF-κB activation in vitro. Using recombinant human enzymes, GYY4137 inhibited the activity of COX-2, iNOS and TNF-α converting enzyme (TACE). In the CFA-treated mouse, GYY4137 (50 mg/kg, i.p.) injected 1 hr prior to CFA increased knee joint swelling while an anti-inflammatory effect, as demonstrated by reduced synovial fluid myeloperoxidase (MPO) and N-acetyl-β-D-glucosaminidase (NAG) activity and decreased TNF-α, IL-1β, IL-6 and IL-8 concentration, was apparent when GYY4137 was injected 6 hrs after CFA. GYY4137 was also anti-inflammatory when given 18 hrs after CFA. Thus, although GYY4137 consistently reduced the generation of pro-inflammatory mediators from human joint cells in vitro, its effect on acute joint inflammation in vivo depended on the timing of administration.
Collapse
Affiliation(s)
- Ling Li
- Pharmaceutical Science Research Division, King's College London, London, England
| | | | | | | | | | | | | | | |
Collapse
|
64
|
Tang XQ, Fang HR, Zhou CF, Zhuang YY, Zhang P, Gu HF, Hu B. A novel mechanism of formaldehyde neurotoxicity: inhibition of hydrogen sulfide generation by promoting overproduction of nitric oxide. PLoS One 2013; 8:e54829. [PMID: 23359814 PMCID: PMC3554621 DOI: 10.1371/journal.pone.0054829] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 12/17/2012] [Indexed: 12/29/2022] Open
Abstract
Background Formaldehyde (FA) induces neurotoxicity by overproduction of intracellular reactive oxygen species (ROS). Increasing studies have shown that hydrogen sulfide (H2S), an endogenous gastransmitter, protects nerve cells against oxidative stress by its antioxidant effect. It has been shown that overproduction of nitric oxide (NO) inhibits the activity of cystathionine-beta-synthase (CBS), the predominant H2S-generating enzyme in the central nervous system. Objective We hypothesize that FA-caused neurotoxicity involves the deficiency of this endogenous protective antioxidant gas, which results from excessive generation of NO. The aim of this study is to evaluate whether FA disturbs H2S synthesis in PC12 cells, and whether this disturbance is associated with overproduction of NO. Principal Findings We showed that exposure of PC12 cells to FA causes reduction of viability, inhibition of CBS expression, decrease of endogenous H2S production, and NO production. CBS silencing deteriorates FA-induced decreases in endogenous H2S generation, neurotoxicity, and intracellular ROS accumulation in PC12 cells; while ADMA, a specific inhibitor of NOS significantly attenuates FA-induced decreases in endogenous H2S generation, neurotoxicity, and intracellular ROS accumulation in PC12 cells. Conclusion/Significance Our data indicate that FA induces neurotoxicity by inhibiting the generation of H2S through excess of NO and suggest that strategies to manipulate endogenous H2S could open a suitable novel therapeutic avenue for FA-induced neurotoxicity.
Collapse
Affiliation(s)
- Xiao-Qing Tang
- Department of Physiology, Medical College, University of South China, Hengyang, Hunan, P. R. China
- * E-mail: (X-QT); (PZ)
| | - Heng-Rong Fang
- Department of Physiology, Medical College, University of South China, Hengyang, Hunan, P. R. China
- Department of Pharmacy, Hengyang Central Hospital, Hengyang, Hunan, P. R. China
| | - Cheng-Fang Zhou
- Department of Physiology, Medical College, University of South China, Hengyang, Hunan, P. R. China
| | - Yuan-Yuan Zhuang
- Department of Physiology, Medical College, University of South China, Hengyang, Hunan, P. R. China
| | - Ping Zhang
- Department of Neurology, Nanhua Affiliated Hospital, University of South China, Hengyang, Hunan, P. R. China
- * E-mail: (X-QT); (PZ)
| | - Hong-Feng Gu
- Department of Physiology, Medical College, University of South China, Hengyang, Hunan, P. R. China
| | - Bi Hu
- Department of Physiology, Medical College, University of South China, Hengyang, Hunan, P. R. China
| |
Collapse
|
65
|
Selenium- and tellurium-containing redox modulators with distinct activity against macrophages: possible implications for the treatment of inflammatory diseases. Tetrahedron 2012. [DOI: 10.1016/j.tet.2012.09.021] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
66
|
Bir SC, Kolluru GK, McCarthy P, Shen X, Pardue S, Pattillo CB, Kevil CG. Hydrogen sulfide stimulates ischemic vascular remodeling through nitric oxide synthase and nitrite reduction activity regulating hypoxia-inducible factor-1α and vascular endothelial growth factor-dependent angiogenesis. J Am Heart Assoc 2012; 1:e004093. [PMID: 23316304 PMCID: PMC3541625 DOI: 10.1161/jaha.112.004093] [Citation(s) in RCA: 125] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2012] [Accepted: 08/14/2012] [Indexed: 12/03/2022]
Abstract
Background Hydrogen sulfide (H2S) therapy is recognized as a modulator of vascular function during tissue ischemia with the notion of potential interactions of nitric oxide (NO) metabolism. However, little is known about specific biochemical mechanisms or the importance of H2S activation of NO metabolism during ischemic tissue vascular remodeling. The goal of this study was to determine the effect of H2S on NO metabolism during chronic tissue ischemia and subsequent effects on ischemic vascular remodeling responses. Methods and Results The unilateral, permanent femoral artery ligation model of hind‐limb ischemia was performed in C57BL/6J wild‐type and endothelial NO synthase–knockout mice to evaluate exogenous H2S effects on NO bioavailability and ischemic revascularization. We found that H2S selectively restored chronic ischemic tissue function and viability by enhancing NO production involving both endothelial NO synthase and nitrite reduction mechanisms. Importantly, H2S increased ischemic tissue xanthine oxidase activity, hind‐limb blood flow, and angiogenesis, which were blunted by the xanthine oxidase inhibitor febuxostat. H2S treatment increased ischemic tissue and endothelial cell hypoxia‐inducible factor‐1α expression and activity and vascular endothelial growth factor protein expression and function in a NO‐dependent manner that was required for ischemic vascular remodeling. Conclusions These data demonstrate that H2S differentially regulates NO metabolism during chronic tissue ischemia, highlighting novel biochemical pathways to increase NO bioavailability for ischemic vascular remodeling.
Collapse
Affiliation(s)
- Shyamal C Bir
- Departments of Pathology and Medicine, LSU Health Sciences Center-Shreveport, Shreveport, LA 71130, USA
| | | | | | | | | | | | | |
Collapse
|
67
|
Wang R. Physiological implications of hydrogen sulfide: a whiff exploration that blossomed. Physiol Rev 2012; 92:791-896. [PMID: 22535897 DOI: 10.1152/physrev.00017.2011] [Citation(s) in RCA: 1416] [Impact Index Per Article: 108.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The important life-supporting role of hydrogen sulfide (H(2)S) has evolved from bacteria to plants, invertebrates, vertebrates, and finally to mammals. Over the centuries, however, H(2)S had only been known for its toxicity and environmental hazard. Physiological importance of H(2)S has been appreciated for about a decade. It started by the discovery of endogenous H(2)S production in mammalian cells and gained momentum by typifying this gasotransmitter with a variety of physiological functions. The H(2)S-catalyzing enzymes are differentially expressed in cardiovascular, neuronal, immune, renal, respiratory, gastrointestinal, reproductive, liver, and endocrine systems and affect the functions of these systems through the production of H(2)S. The physiological functions of H(2)S are mediated by different molecular targets, such as different ion channels and signaling proteins. Alternations of H(2)S metabolism lead to an array of pathological disturbances in the form of hypertension, atherosclerosis, heart failure, diabetes, cirrhosis, inflammation, sepsis, neurodegenerative disease, erectile dysfunction, and asthma, to name a few. Many new technologies have been developed to detect endogenous H(2)S production, and novel H(2)S-delivery compounds have been invented to aid therapeutic intervention of diseases related to abnormal H(2)S metabolism. While acknowledging the challenges ahead, research on H(2)S physiology and medicine is entering an exponential exploration era.
Collapse
Affiliation(s)
- Rui Wang
- Department of Biology, Lakehead University, Thunder Bay, Ontario, Canada.
| |
Collapse
|
68
|
Role of endogenous hydrogen sulfide in neurogenic relaxation of rat corpus cavernosum. Biochem Pharmacol 2012; 83:1261-8. [DOI: 10.1016/j.bcp.2012.01.026] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2011] [Revised: 01/19/2012] [Accepted: 01/19/2012] [Indexed: 12/11/2022]
|
69
|
Predmore BL, Julian D, Cardounel AJ. Hydrogen sulfide increases nitric oxide production from endothelial cells by an akt-dependent mechanism. Front Physiol 2011; 2:104. [PMID: 22194727 PMCID: PMC3242361 DOI: 10.3389/fphys.2011.00104] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2011] [Accepted: 12/01/2011] [Indexed: 12/22/2022] Open
Abstract
Hydrogen sulfide (H2S) and nitric oxide (NO) are both gasotransmitters that can elicit synergistic vasodilatory responses in the in the cardiovascular system, but the mechanisms behind this synergy are unclear. In the current study we investigated the molecular mechanisms through which H2S regulates endothelial NO production. Initial studies were performed to establish the temporal and dose-dependent effects of H2S on NO generation using EPR spin trapping techniques. H2S stimulated a twofold increase in NO production from endothelial nitric oxide synthase (eNOS), which was maximal 30 min after exposure to 25–150 μM H2S. Following 30 min H2S exposure, eNOS phosphorylation at Ser 1177 was significantly increased compared to control, consistent with eNOS activation. Pharmacological inhibition of Akt, the kinase responsible for Ser 1177 phosphorylation, attenuated the stimulatory effect of H2S on NO production. Taken together, these data demonstrate that H2S up-regulates NO production from eNOS through an Akt-dependent mechanism. These results implicate H2S in the regulation of NO production in endothelial cells, and suggest that deficiencies in H2S signaling can directly impact processes regulated by NO.
Collapse
|
70
|
Kurokawa Y, Sekiguchi F, Kubo S, Yamasaki Y, Matsuda S, Okamoto Y, Sekimoto T, Fukatsu A, Nishikawa H, Kume T, Fukushima N, Akaike A, Kawabata A. Involvement of ERK in NMDA receptor-independent cortical neurotoxicity of hydrogen sulfide. Biochem Biophys Res Commun 2011; 414:727-32. [PMID: 22001931 DOI: 10.1016/j.bbrc.2011.09.144] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Accepted: 09/28/2011] [Indexed: 11/30/2022]
Abstract
Hydrogen sulfide (H(2)S), a gasotransmitter, exerts both neurotoxicity and neuroprotection, and targets multiple molecules including NMDA receptors, T-type calcium channels and NO synthase (NOS) that might affect neuronal viability. Here, we determined and characterized effects of NaHS, an H(2)S donor, on cell viability in the primary cultures of mouse fetal cortical neurons. NaHS caused neuronal death, as assessed by LDH release and trypan blue staining, but did not significantly reduce the glutamate toxicity. The neurotoxicity of NaHS was resistant to inhibitors of NMDA receptors, T-type calcium channels and NOS, and was blocked by inhibitors of MEK, but not JNK, p38 MAP kinase, PKC and Src. NaHS caused prompt phosphorylation of ERK and upregulation of Bad, followed by translocation of Bax to mitochondria and release of mitochondrial cytochrome c, leading to the nuclear condensation/fragmentation. These effects of NaHS were suppressed by the MEK inhibitor. Our data suggest that the NMDA receptor-independent neurotoxicity of H(2)S involves activation of the MEK/ERK pathway and some apoptotic mechanisms.
Collapse
Affiliation(s)
- Yuko Kurokawa
- Division of Pharmacology and Pathophysiology, Kinki University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Félétou M. The Endothelium, Part I: Multiple Functions of the Endothelial Cells -- Focus on Endothelium-Derived Vasoactive Mediators. ACTA ACUST UNITED AC 2011. [DOI: 10.4199/c00031ed1v01y201105isp019] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
72
|
|
73
|
Effects of intravenous sulfide during porcine aortic occlusion-induced kidney ischemia/reperfusion injury. Shock 2011; 35:156-63. [PMID: 20661185 DOI: 10.1097/shk.0b013e3181f0dc91] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
In rodents, inhaled H2S and injection of H2S donors protected against kidney ischemia/reperfusion (I/R) injury. During porcine aortic occlusion, the H2S donor Na2S (sulfide) reduced energy expenditure and decreased the noradrenaline requirements needed to maintain hemodynamic targets during early reperfusion. Therefore, we tested the hypothesis whether sulfide pretreatment may also ameliorate organ function in porcine aortic occlusion-induced kidney I/R injury. Anesthetized, ventilated, and instrumented pigs randomly received either sulfide or vehicle and underwent 90 min of kidney ischemia using intraaortic balloon-occlusion, and 8 h of reperfusion. During reperfusion, noradrenaline was titrated to maintain blood pressure at baseline levels. Sulfide attenuated the fall in creatinine clearance and the rise in creatinine blood levels, whereas renal blood flow and fractional Na+ excretion were comparable. Sulfide also lowered the blood IL-6, IL-1β, and nitrite + nitrate concentrations, which coincided with reduced kidney oxidative DNA base damage and iNOS expression, and attenuated glomerular histological injury as assessed by the incidence of glomerular tubularization. While expression of heme oxygenase 1 and cleaved caspase 3 did not differ, sulfide reduced the expression Bcl-xL and increased the activation of nuclear transcription factor κB. During porcine aortic occlusion-induced kidney I/R injury, sulfide pretreatment attenuated tissue injury and organ dysfunction as a result of reduced inflammation and oxidative and nitrosative stress. The higher nuclear transcription factor κB activation was probably due to the drop in temperature.
Collapse
|
74
|
Liaw RL, Srilatha B, Adaikan PG. Effects of hydrogen sulfide on erectile function and its possible mechanism(s) of action. J Sex Med 2011; 8:1853-64. [PMID: 21492403 DOI: 10.1111/j.1743-6109.2011.02279.x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
INTRODUCTION The current pharmacotherapy for erectile dysfunction (ED) relies significantly on the use of phosphodiesterase type 5 (PDE5) inhibitors, but quite a proportion of ED patients are resistant to this therapy, necessitating a search for an alternative treatment. We reviewed available published data to analyze current evidence of hydrogen sulfide (H(2) S) as a novel pharmacotherapeutic agent with supportive role in sexual function. AIM To discuss the role of H(2) S in erectile function, its possible mechanism of action, and how this knowledge may be exploited for therapeutic use. METHODS Pubmed and Medline search was conducted to identify original articles and reviews. MAIN OUTCOME MEASURES Data from peer-reviewed publications. RESULTS Animal studies using different species, including in vitro study done in humans, show evidence of H(2) S's pro-erectile effects. The mechanism behind is still unclear, but evidence in literature points out the involvement of K(+) (ATP) channel, modulation of protein with anti-erectile effects, as well as involvement of the nitrergic pathway through a complex cross-talk. A new drug called H(2) S-donating sildenafil (ACS6), which incorporated an H(2) S-donating moiety in sildenafil, has been developed. While more studies are still needed, this heralded a new pharmacotherapeutical approach, which is multipronged in nature. CONCLUSIONS Given the mounting evidence of H(2) S's role in erectile function and how it appears to achieve its pro-erectile effects through different mechanisms, H(2) S represents a potentially important treatment alternative or adjunct to PDE5 inhibitors.
Collapse
Affiliation(s)
- Roeswita Leono Liaw
- Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University Hospital, National University of Singapore, Singapore
| | | | | |
Collapse
|
75
|
Hydrogen sulfide-mediated cardioprotection: mechanisms and therapeutic potential. Clin Sci (Lond) 2010; 120:219-29. [DOI: 10.1042/cs20100462] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
H2S (hydrogen sulfide), viewed with dread for more than 300 years, is rapidly becoming a ubiquitously present and physiologically relevant signalling molecule. Knowledge of the production and metabolism of H2S has spurred interest in delineating its functions both in physiology and pathophysiology of disease. Although its role in blood pressure regulation and interaction with NO is controversial, H2S, through its anti-apoptotic, anti-inflammatory and antioxidant effects, has demonstrated significant cardioprotection. As a result, a number of sulfide-donor drugs, including garlic-derived polysulfides, are currently being designed and investigated for the treatment of cardiovascular conditions, specifically myocardial ischaemic disease. However, huge gaps remain in our knowledge about this gasotransmitter. Only by additional studies will we understand more about the role of this intriguing molecule in the treatment of cardiovascular disease.
Collapse
|
76
|
Lisjak M, Srivastava N, Teklic T, Civale L, Lewandowski K, Wilson I, Wood ME, Whiteman M, Hancock JT. A novel hydrogen sulfide donor causes stomatal opening and reduces nitric oxide accumulation. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2010; 48:931-5. [PMID: 20970349 DOI: 10.1016/j.plaphy.2010.09.016] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2010] [Revised: 09/21/2010] [Accepted: 09/22/2010] [Indexed: 05/03/2023]
Abstract
Effects of hydrogen sulfide (H(2)S) on plant physiology have been previously studied, but such studies have relied on the use of NaSH as a method for supplying H(2)S to tissues. Now new compounds which give a less severe H(2)S shock and a more prolonged exposure to H(2)S have been developed. Here the effects of one such compound, GYY4137, has been investigated to determine its effects on stomatal closure in Arabidopsis thaliana. It was found that both NaSH and GYY4137 caused stomatal opening in the light and prevented stomatal closure in the dark. Nitric oxide (NO) has been well established as a mediator of stomatal movements and here it was found that both NaSH and GYY4137 reduced the accumulation of NO in guard cells, perhaps suggesting a mode of action for H(2)S in this system. GYY4137, and future related compounds, will be important tools to unravel the effects of plant exposure to H(2)S and to determine how H(2)S may fit into plant cell signalling pathways.
Collapse
Affiliation(s)
- M Lisjak
- Department of Agroecology, University of J. J. Strossmayer, Osijek, Croatia
| | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Curative effects of hydrogen sulfide against acetaminophen-induced hepatotoxicity in mice. Life Sci 2010; 87:692-8. [PMID: 20951146 DOI: 10.1016/j.lfs.2010.10.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2010] [Revised: 09/13/2010] [Accepted: 10/06/2010] [Indexed: 11/20/2022]
Abstract
AIMS Hydrogen sulfide (H(2)S), an endogenous gaseous mediator, plays an important role in regulation of many physiological and pathological processes. On the other hand, acetaminophen overdose is a major cause of drug-induced liver failure. The aim of this study therefore is to evaluate the possible curative effects of H(2)S against acetaminophen-induced hepatotoxicity. MAIN METHODS Male Swiss mice were treated with sodium hydrogen sulfide, a H(2)S donor, 30 min after acetaminophen administration. N-acetylcysteine, a therapeutic antidote, was used as a reference drug. KEY FINDINGS H(2)S treatment resulted in hepatocurative effects as evident by a significant decrease in serum alanine aminotransferase and hepatic malondialdehyde and nitric oxide levels, with a concurrent increase in hepatic glutathione content compared to acetaminophen-treated group. H(2)S did not alter catalase activity. Additionally, immunohistochemical analysis demonstrated that H(2)S treatment markedly reduced tumor necrosis factor-α expression, while expression of cyclooxygenase-2 was markedly enhanced with nuclear localization into hepatocytes. The curative effects of H(2)S were confirmed by liver histopathological examination and were maintained in the presence of glibenclamide, an antagonist of ATP-sensitive potassium (K(ATP)) channels. SIGNIFICANCE H(2)S treatment markedly alleviates acetaminophen hepatotoxicity in mice possibly, in part, through anti-oxidative and anti-inflammatory effects but not likely to be coupled with activation of K(ATP) channels. The hepatocurative effects of H(2)S are comparable to N-acetylcysteine. Hence, H(2)S has a potential therapeutic value for treatment of acetaminophen hepatotoxicity.
Collapse
|
78
|
Ganster F, Burban M, de la Bourdonnaye M, Fizanne L, Douay O, Loufrani L, Mercat A, Calès P, Radermacher P, Henrion D, Asfar P, Meziani F. Effects of hydrogen sulfide on hemodynamics, inflammatory response and oxidative stress during resuscitated hemorrhagic shock in rats. CRITICAL CARE : THE OFFICIAL JOURNAL OF THE CRITICAL CARE FORUM 2010; 14:R165. [PMID: 20836847 PMCID: PMC3219260 DOI: 10.1186/cc9257] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Revised: 05/15/2010] [Accepted: 09/13/2010] [Indexed: 01/15/2023]
Abstract
Introduction Hydrogen sulfide (H2S) has been shown to improve survival in rodent models of lethal hemorrhage. Conversely, other authors have reported that inhibition of endogenous H2S production improves hemodynamics and reduces organ injury after hemorrhagic shock. Since all of these data originate from unresuscitated models and/or the use of a pre-treatment design, we therefore tested the hypothesis that the H2S donor, sodium hydrosulfide (NaHS), may improve hemodynamics in resuscitated hemorrhagic shock and attenuate oxidative and nitrosative stresses. Methods Thirty-two rats were mechanically ventilated and instrumented to measure mean arterial pressure (MAP) and carotid blood flow (CBF). Animals were bled during 60 minutes in order to maintain MAP at 40 ± 2 mm Hg. Ten minutes prior to retransfusion of shed blood, rats randomly received either an intravenous bolus of NaHS (0.2 mg/kg) or vehicle (0.9% NaCl). At the end of the experiment (T = 300 minutes), blood, aorta and heart were harvested for Western blot (inductible Nitric Oxyde Synthase (iNOS), Nuclear factor-κB (NF-κB), phosphorylated Inhibitor κB (P-IκB), Inter-Cellular Adhesion Molecule (I-CAM), Heme oxygenase 1(HO-1), Heme oxygenase 2(HO-2), as well as nuclear respiratory factor 2 (Nrf2)). Nitric oxide (NO) and superoxide anion (O2-) were also measured by electron paramagnetic resonance. Results At the end of the experiment, control rats exhibited a decrease in MAP which was attenuated by NaHS (65 ± 32 versus 101 ± 17 mmHg, P < 0.05). CBF was better maintained in NaHS-treated rats (1.9 ± 1.6 versus 4.4 ± 1.9 ml/minute P < 0.05). NaHS significantly limited shock-induced metabolic acidosis. NaHS also prevented iNOS expression and NO production in the heart and aorta while significantly reducing NF-kB, P-IκB and I-CAM in the aorta. Compared to the control group, NaHS significantly increased Nrf2, HO-1 and HO-2 and limited O2- release in both aorta and heart (P < 0.05). Conclusions NaHS is protective against the effects of ischemia reperfusion induced by controlled hemorrhage in rats. NaHS also improves hemodynamics in the early resuscitation phase after hemorrhagic shock, most likely as a result of attenuated oxidative stress. The use of NaHS hence appears promising in limiting the consequences of ischemia reperfusion (IR).
Collapse
Affiliation(s)
- Frédérique Ganster
- Laboratoire HIFIH, UPRES EA 3859, IFR 132, Université d'Angers, Rue Haute de Reculée, Angers, F-49035 France.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
79
|
Takahashi T, Aoki Y, Okubo K, Maeda Y, Sekiguchi F, Mitani K, Nishikawa H, Kawabata A. Upregulation of Ca(v)3.2 T-type calcium channels targeted by endogenous hydrogen sulfide contributes to maintenance of neuropathic pain. Pain 2010; 150:183-191. [PMID: 20546998 DOI: 10.1016/j.pain.2010.04.022] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2009] [Revised: 04/09/2010] [Accepted: 04/20/2010] [Indexed: 11/28/2022]
Abstract
Hydrogen sulfide (H(2)S) formed from l-cysteine by multiple enzymes including cystathionine-gamma-lyase (CSE) is now considered a gasotransmitter in the mammalian body. Our previous studies have shown that H(2)S activates/sensitizes Ca(v)3.2 T-type Ca(2+) channels, leading to facilitation of somatic and visceral nociception, and that CSE-derived endogenous H(2)S participates in inflammatory pain. Here, we show novel evidence for involvement of the endogenous H(2)S-Ca(v)3.2 pathway in neuropathic pain. In the rat subjected to the right L5 spinal nerve cutting (L5SNC), a neuropathic pain model, i.p. administration of dl-propargylglycine (PPG) and beta-cyanoalanine, irreversible and reversible CSE inhibitors, respectively, strongly suppressed the neuropathic hyperalgesia/allodynia. The anti-hyperalgesic effect of PPG was reversed by intraplantar administration of NaHS, a donor for H(2)S, in the L5SNC rat. Intraplantar administration or topical application of mibefradil, a T-type Ca(2+) channel blocker, reversed hyperalgesia in the L5SNC rat. The protein levels of Ca(v)3.2, but not CSE, in the ipsilateral L4, L5 and L6 dorsal root ganglia were dramatically upregulated in the L5SNC rat. Finally, silencing of Ca(v)3.2 in DRG by repeated intrathecal administration of Ca(v)3.2-targeting siRNA significantly attenuated the neuropathic hyperalgesia in the L5SNC rat. In conclusion, our data suggest that Ca(v)3.2 T-type Ca(2+) channels in sensory neurons are upregulated and activated/sensitized by CSE-derived endogenous H(2)S after spinal nerve injury, contributing to the maintenance of neuropathic pain. We thus propose that Ca(v)3.2 and CSE could be targets for the development of therapeutic drugs for the treatment of neuropathic pain.
Collapse
Affiliation(s)
- Tomoko Takahashi
- Division of Pharmacology & Pathophysiology, Kinki University School of Pharmacy, Higashi-Osaka 577-8502, Japan
| | | | | | | | | | | | | | | |
Collapse
|
80
|
Ekundi-Valentim E, Santos KT, Camargo EA, Denadai-Souza A, Teixeira SA, Zanoni CI, Grant AD, Wallace JL, Muscará MN, Costa SK. Differing effects of exogenous and endogenous hydrogen sulphide in carrageenan-induced knee joint synovitis in the rat. Br J Pharmacol 2010; 159:1463-74. [PMID: 20136840 PMCID: PMC2850403 DOI: 10.1111/j.1476-5381.2010.00640.x] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2009] [Revised: 11/05/2009] [Accepted: 11/13/2009] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND AND PURPOSE Recent findings suggest that the noxious gas H(2)S is produced endogenously, and that physiological concentrations of H(2)S are able to modulate pain and inflammation in rodents. This study was undertaken to evaluate the ability of endogenous and exogenous H(2)S to modulate carrageenan-induced synovitis in the rat knee. EXPERIMENTAL APPROACH Synovitis was induced in Wistar rats by intra-articular injection of carrageenan into the knee joint. Sixty minutes prior to carrageenan injection, the rats were pretreated with indomethacin, an inhibitor of H(2)S formation (DL-propargylglycine) or an H(2)S donor [Lawesson's reagent (LR)]. KEY RESULTS Injection of carrageenan evoked knee inflammation, pain as characterized by impaired gait, secondary tactile allodynia of the ipsilateral hindpaw, joint swelling, histological changes, inflammatory cell infiltration, increased synovial myeloperoxidase, protein nitrotyrosine residues, inducible NOS (iNOS) activity and NO production. Pretreatment with LR or indomethacin significantly attenuated the pain responses, and all the inflammatory and biochemical changes, except for the increased iNOS activity, NO production and 3-NT. Propargylglycine pretreatment potentiated synovial iNOS activity (and NO production), and enhanced macrophage infiltration, but had no effect on other inflammatory parameters. CONCLUSIONS AND IMPLICATIONS Whereas exogenous H(2)S delivered to the knee joint can produce a significant anti-inflammatory and anti-nociceptive effect, locally produced H(2)S exerts little immunomodulatory effect. These data further support the development and use of H(2)S donors as potential alternatives (or complementary therapies) to the available anti-inflammatory compounds used for treatment of joint inflammation or relief of its symptoms.
Collapse
Affiliation(s)
- E Ekundi-Valentim
- Department of Pharmacology, Institute of Biomedical Sciences, University of São PauloSão Paulo, SP, Brazil
| | - KT Santos
- Department of Pharmacology, Institute of Biomedical Sciences, University of São PauloSão Paulo, SP, Brazil
| | - EA Camargo
- Department of Pharmacology, Institute of Biomedical Sciences, University of São PauloSão Paulo, SP, Brazil
- Department of Physiology, Federal University of SergipeAracaju-SE, Brazil
| | - A Denadai-Souza
- Department of Pharmacology, Institute of Biomedical Sciences, University of São PauloSão Paulo, SP, Brazil
| | - SA Teixeira
- Department of Pharmacology, Institute of Biomedical Sciences, University of São PauloSão Paulo, SP, Brazil
| | - CI Zanoni
- Department of Pharmacology, Institute of Biomedical Sciences, University of São PauloSão Paulo, SP, Brazil
| | - AD Grant
- Wolfson Centre for Age-related Diseases, King's CollegeLondon, UK
| | - JL Wallace
- Farncombe Institute, Department of Medicine, McMaster UniversityHamilton, ON, Canada
| | - MN Muscará
- Department of Pharmacology, Institute of Biomedical Sciences, University of São PauloSão Paulo, SP, Brazil
| | - SK Costa
- Department of Pharmacology, Institute of Biomedical Sciences, University of São PauloSão Paulo, SP, Brazil
| |
Collapse
|
81
|
Zhang H, Tang J, Liu XP, Wang Y, Yu W, Peng WY, Fang F, Ma DF, Wei ZJ, Hu LY. Hydrogen sulfide promotes root organogenesis in Ipomoea batatas, Salix matsudana and Glycine max. JOURNAL OF INTEGRATIVE PLANT BIOLOGY 2009; 51:1086-94. [PMID: 20021556 DOI: 10.1111/j.1744-7909.2009.00885.x] [Citation(s) in RCA: 129] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
In this report, we demonstrate that sodium hydrosulfide (NaHS), a hydrogen sulfide (H(2)S) donor, promoted adventitious root formation mediated by auxin and nitric oxide (NO). Application of the H(2)S donor to seedling cuttings of sweet potato (Ipomoea batatas L.) promoted the number and length of adventitious roots in a dose-dependent manner. It was also verified that H(2)S or HS(-) rather than other sulfur-containing components derived from NaHS could be attributed to the stimulation of adventitious root formation. A rapid increase in endogenous H(2)S, indole acetic acid (IAA) and NO were sequentially observed in shoot tips of sweet potato seedlings treated with HaHS. Further investigation showed that H(2)S-mediated root formation was alleviated by N-1-naphthylphthalamic acid (NPA), an IAA transport inhibitor, and 2-(4-carboxyphenyl)-4,4,5,5-tetramethylimidazoline-1-oxyl-3-oxide (cPTIO), an NO scavenger. Similar phenomena in H(2)S donor-dependent root organogenesis were observed in both excised willow (Salix matsudana var. tortuosa Vilm) shoots and soybean (Glycine max L.) seedlings. These results indicated that the process of H(2)S-induced adventitious root formation was likely mediated by IAA and NO, and that H(2)S acts upstream of IAA and NO signal transduction pathways.
Collapse
Affiliation(s)
- Hua Zhang
- School of Biotechnology and Food Engineering, Hefei University of Technology, Hefei 230009, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Whiteman M, Moore PK. Hydrogen sulfide and the vasculature: a novel vasculoprotective entity and regulator of nitric oxide bioavailability? J Cell Mol Med 2009; 13:488-507. [PMID: 19374684 PMCID: PMC3822510 DOI: 10.1111/j.1582-4934.2009.00645.x] [Citation(s) in RCA: 220] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Hydrogen sulfide (H2S) is a well known and pungent toxic gas that has recently been shown to be synthesised in man from the amino acids cystathionine, homocysteine and cysteine by at least two distinct enzymes; cystathionine-γ-lyase and cystathionine-β-synthase. In the past few years, H2S has emerged as a novel and increasingly important mediator in the cardiovascular system but delineating the precise physiology and pathophysiology of H2S is proving to be complex and difficult to unravel with disparate findings reported with cell types, tissue types and animal species reported. Therefore, in this review we summarize the mechanisms by which H2S has been proposed to regulate blood pressure and cardiac function, discuss the mechanistic discrepancies reported in the literature as well as the therapeutic potential of H2S. We also examine the methods of H2S detection in biological fluids, processes for H2S removal and discuss the reported blood levels of H2S in man and animal models of cardiovascular pathology. We also highlight the complex interaction of H2S with nitric oxide in regulating cardiovascular function in health and disease.
Collapse
Affiliation(s)
- Matthew Whiteman
- Institute of Biomedical and Clinical Science, Peninsula Medical School, St Luke's Campus, Exeter, UK.
| | | |
Collapse
|
83
|
Li L, Hsu A, Moore PK. Actions and interactions of nitric oxide, carbon monoxide and hydrogen sulphide in the cardiovascular system and in inflammation--a tale of three gases! Pharmacol Ther 2009; 123:386-400. [PMID: 19486912 DOI: 10.1016/j.pharmthera.2009.05.005] [Citation(s) in RCA: 240] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Accepted: 05/15/2009] [Indexed: 01/17/2023]
Abstract
Nitric oxide (NO), carbon monoxide (CO) and hydrogen sulphide (H(2)S) together make up a family of biologically active gases (the so-called 'gaseous triumvirate') with an increasingly well defined range of physiological effects plus roles to play in a number of disease states. Over the years, most researchers have concentrated their attention on understanding the part played by a single gas in one or more body systems. It is becoming more clear that all three gases are synthesised naturally in the body, often by the same cells within the same organs, and that all three gases exert essentially similar biological effects albeit via different mechanisms. Within the cardiovascular system, for example, all are vasodilators, promote angiogenesis and vascular remodelling and are protective towards tissue damage in for example, ischaemia-reperfusion injury in the heart. Similarly, all exhibit complex effects in inflammation with both pro- and anti-inflammatory effects recognised. It seems likely that cell function is controlled not by the activity of single gases working in isolation but by the concerted activity of all three of these gases working together.
Collapse
Affiliation(s)
- Ling Li
- Pharmaceutical Science Division, King's College London, Franklin-Wilkins Building, 150 Stamford Street, London SE19NH, UK
| | | | | |
Collapse
|
84
|
Maeda Y, Aoki Y, Sekiguchi F, Matsunami M, Takahashi T, Nishikawa H, Kawabata A. Hyperalgesia induced by spinal and peripheral hydrogen sulfide: evidence for involvement of Cav3.2 T-type calcium channels. Pain 2009; 142:127-32. [PMID: 19167819 DOI: 10.1016/j.pain.2008.12.021] [Citation(s) in RCA: 110] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2008] [Revised: 11/20/2008] [Accepted: 12/15/2008] [Indexed: 11/24/2022]
Abstract
Hydrogen sulfide (H2S), a gasotransmitter, facilitates membrane currents through T-type Ca2+ channels, and intraplantar (i.pl.) administration of NaHS, a donor of H2S, causes prompt hyperalgesia in rats. In this context, we asked whether intrathecal (i.t.) administration of NaHS could mimic the hyperalgesic effect of i.pl. NaHS in rats, and then examined if Cav3.2 isoform of T-type Ca2+ channels contributed to the pro-nociceptive effects of i.t. and i.pl. NaHS. Either i.t. or i.pl. administration of NaHS rapidly decreased nociceptive threshold in rats, as determined by the paw pressure method. The hyperalgesia caused by i.t. and i.pl. NaHS was abolished by co-administration of mibefradil, a pan-T-type Ca2+ channel inhibitor, and also suppressed by pretreatment with i.t. and i.pl. zinc chloride, known to preferentially inhibit Cav3.2 among T-type Ca2+ channel isoforms, respectively. Repeated i.t. administration of antisense oligodeoxynucleotides (ODNs) targeting rat Cav3.2, but not mismatch ODNs, caused silencing of Cav3.2 protein in the dorsal root ganglia and spinal cord, and then attenuated the hyperalgesia induced by either i.t. or i.pl. NaHS. Our findings thus establish that spinal and peripheral NaHS/H2S activates or sensitizes Cav3.2 T-type Ca2+ channels expressed in the primary afferents and/or spinal nociceptive neurons, leading to sensitization of nociceptive processing and hyperalgesia.
Collapse
Affiliation(s)
- Yumi Maeda
- Division of Pharmacology and Pathophysiology, Kinki University School of Pharmacy, 3-4-1 Kowakae, Higashi-Osaka 577-8502, Japan
| | | | | | | | | | | | | |
Collapse
|
85
|
Hydrogen sulfide decreases adenosine triphosphate levels in aortic rings and leads to vasorelaxation via metabolic inhibition. Life Sci 2008; 83:589-94. [PMID: 18790700 DOI: 10.1016/j.lfs.2008.08.006] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2008] [Revised: 07/04/2008] [Accepted: 08/15/2008] [Indexed: 12/16/2022]
Abstract
AIMS Hydrogen sulfide (H(2)S) at low concentrations serves as a physiological endogenous vasodilator molecule, while at higher concentrations it can trigger cytotoxic effects. The aim of our study was to elucidate the potential mechanisms responsible for the effects of H(2)S on vascular tone. MAIN METHODS We measured the vascular tone in vitro in precontracted rat thoracic aortic rings and we have tested the effect of different oxygen levels and a variety of inhibitors affecting known vasodilatory pathways. We have also compared the vascular effect of high concentrations of H(2)S to those of pharmacological inhibitors of oxidative phosphorylation. Furthermore, we measured adenosine triphosphate (ATP)-levels in the same vascular tissues. KEY FINDINGS We have found that in rat aortic rings: (1) H(2)S decreases ATP levels; (2) relaxations to H(2)S depend on the ambient oxygen concentration; (3) prostaglandins do not take part in the H(2)S induced relaxations; (4) the 3':5'-cyclic guanosine monophosphate (cGMP)-nitric oxide (NO) pathway does not have a role in the relaxations (5) the role of K(ATP) channels is limited, while Cl(-)/HCO(3)(-) channels have a role in the relaxations. (6): We have observed that high concentrations of H(2)S relax the aortic rings in a fashion similar to sodium cyanide, and both agents reduce cellular ATP levels to a comparable degree. SIGNIFICANCE H(2)S, a new gasotransmitter of emerging importance, leads to relaxation via Cl(-)/HCO(3)(-) channels and metabolic inhibition and the interactions of these two factors depend on the oxygen levels of the tissue.
Collapse
|