51
|
Sakamoto K, Suzuki Y, Kurauchi Y, Mori A, Nakahara T, Ishii K. Hydrogen sulfide attenuates NMDA-induced neuronal injury via its anti-oxidative activity in the rat retina. Exp Eye Res 2014; 120:90-6. [DOI: 10.1016/j.exer.2014.01.008] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Revised: 01/06/2014] [Accepted: 01/10/2014] [Indexed: 12/29/2022]
|
52
|
Guo C, Liang F, Shah Masood W, Yan X. Hydrogen sulfide protected gastric epithelial cell from ischemia/reperfusion injury by Keap1 s-sulfhydration, MAPK dependent anti-apoptosis and NF-κB dependent anti-inflammation pathway. Eur J Pharmacol 2014; 725:70-8. [PMID: 24444438 DOI: 10.1016/j.ejphar.2014.01.009] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Revised: 12/20/2013] [Accepted: 01/08/2014] [Indexed: 12/15/2022]
Abstract
Hydrogen sulfide (H2S) has been proposed as a novel gas-transmittter, which plays multiple physiological and pathological functions in various body systems, including gastrointestinal tract. The present study was undertaken to investigate the effects and mechanisms of H2S pharmacological preconditioning on gastric epithelial cells ischemia-reperfusion (I/R) injury. We report here that sodium hydrosulfide (NaHS), an H2S donor, concentration-dependently suppressed I/R-induced cellular injury and apoptotic cell death. This protection effect was also confirmed by endogenous over-producing H2S. Furthermore, NaHS also prevented I/R-induced oxidative stress and inflammatory responses, evidenced by increases in GSH level, decreases in MDA contents, reactive oxygen species generation and secretions of NO, IL-6 and TNF-α. NaHS also prevented I/R-induced p38- and c-Jun NH2-terminal kinase (JNK)-mitogen-activated protein kinase (MAPK) phosphorylation and NF-κB activation. H2S also induced Keap1 s-sulfhydration, and further Keap1/Nrf2 disassociation and Nrf2 activation. H2S exerted its protective effect through reactive oxygen species clearance, inhibition of p38 and JNK dependent cell apoptosis and NF-κB dependent inflammation pathway. Our results provide evidence that H2S may have potential therapeutic value in acute gastric mucosal lesion, which is often caused by ischemia/reperfusion.
Collapse
Affiliation(s)
- Cheng Guo
- Department of Hepatobiliary Surgery, First Affiliated Hospital of Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Fenli Liang
- Center for Cancer Research, Medical school, Xi'an Jiaotong University, Xi'an 710061, Shaanxi, China.
| | - Walayat Shah Masood
- Department of Pathology, Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan.
| | - Xiaofei Yan
- Department of Genetics and Molecular Biology, Medical College of Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
53
|
Zhang YY, Li HX, Chen YY, Fang H, Yu YN, Liu J, Jing ZW, Wang Z, Wang YY. Convergent and divergent pathways decoding hierarchical additive mechanisms in treating cerebral ischemia-reperfusion injury. CNS Neurosci Ther 2013; 20:253-63. [PMID: 24351012 DOI: 10.1111/cns.12205] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2013] [Revised: 10/14/2013] [Accepted: 10/15/2013] [Indexed: 12/29/2022] Open
Abstract
INTRODUCTION Cerebral ischemia is considered to be a highly complex disease resulting from the complicated interplay of multiple pathways. Disappointedly, most of the previous studies were limited to a single gene or a single pathway. The extent to which all involved pathways are translated into fusing mechanisms of a combination therapy is of fundamental importance. AIMS We report an integrative strategy to reveal the additive mechanism that a combination (BJ) of compound baicalin (BA) and jasminoidin (JA) fights against cerebral ischemia based on variation of pathways and functional communities. RESULTS We identified six pathways of BJ group that shared diverse additive index from 0.09 to 1, which assembled broad cross talks from seven pathways of BA and 16 pathways of JA both at horizontal and vertical levels. Besides a total of 60 overlapping functions as a robust integration background among the three groups based on significantly differential subnetworks, additive mechanism with strong confidence by networks altered functions. CONCLUSIONS These results provide strong evidence that the additive mechanism is more complex than previously appreciated, and an integrative analysis of pathways may suggest an important paradigm for revealing pharmacological mechanisms underlying drug combinations.
Collapse
Affiliation(s)
- Ying-Ying Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | | | |
Collapse
|
54
|
Liao C, Hua Y. Effect of hydrogen sulphide on the expression of osteoprotegerin and receptor activator of NF-κB ligand in human periodontal ligament cells induced by tension-force stimulation. Arch Oral Biol 2013; 58:1784-90. [DOI: 10.1016/j.archoralbio.2013.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2013] [Revised: 07/08/2013] [Accepted: 08/05/2013] [Indexed: 01/03/2023]
|
55
|
Clanton TL, Hogan MC, Gladden LB. Regulation of cellular gas exchange, oxygen sensing, and metabolic control. Compr Physiol 2013; 3:1135-90. [PMID: 23897683 DOI: 10.1002/cphy.c120030] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cells must continuously monitor and couple their metabolic requirements for ATP utilization with their ability to take up O2 for mitochondrial respiration. When O2 uptake and delivery move out of homeostasis, cells have elaborate and diverse sensing and response systems to compensate. In this review, we explore the biophysics of O2 and gas diffusion in the cell, how intracellular O2 is regulated, how intracellular O2 levels are sensed and how sensing systems impact mitochondrial respiration and shifts in metabolic pathways. Particular attention is paid to how O2 affects the redox state of the cell, as well as the NO, H2S, and CO concentrations. We also explore how these agents can affect various aspects of gas exchange and activate acute signaling pathways that promote survival. Two kinds of challenges to gas exchange are also discussed in detail: when insufficient O2 is available for respiration (hypoxia) and when metabolic requirements test the limits of gas exchange (exercising skeletal muscle). This review also focuses on responses to acute hypoxia in the context of the original "unifying theory of hypoxia tolerance" as expressed by Hochachka and colleagues. It includes discourse on the regulation of mitochondrial electron transport, metabolic suppression, shifts in metabolic pathways, and recruitment of cell survival pathways preventing collapse of membrane potential and nuclear apoptosis. Regarding exercise, the issues discussed relate to the O2 sensitivity of metabolic rate, O2 kinetics in exercise, and influences of available O2 on glycolysis and lactate production.
Collapse
Affiliation(s)
- T L Clanton
- Department of Applied Physiology and Kinesiology, University of Florida, Gainesville, Florida, USA.
| | | | | |
Collapse
|
56
|
Guo W, Cheng ZY, Zhu YZ. Hydrogen sulfide and translational medicine. Acta Pharmacol Sin 2013; 34:1284-91. [PMID: 24096643 PMCID: PMC3791558 DOI: 10.1038/aps.2013.127] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 08/12/2013] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2S) along with carbon monoxide and nitric oxide is an important signaling molecule that has undergone large numbers of fundamental investigations. H2S is involved in various physiological activities associated with the regulation of homeostasis, vascular contractility, pro- and anti-inflammatory activities, as well as pro- and anti-apoptotic activities etc. However, the actions of H2S are influenced by its concentration, reaction time, and cell/disease types. Therefore, H2S is a signaling molecule without definite effect. The use of existing H2S donors is limited because of the instant release and short lifetime of H2S. Thus, translational medicine involving the sustained and controlled release of H2S is of great value for both scientific and clinical uses. H2S donation can be manipulated by different ways, including where H2S is given, how H2S is donated, or the specific structures of H2S-releasing drugs and H2S donor molecules. This review briefly summarizes recent progress in research on the physiological and pathological functions of H2S and H2S-releasing drugs, and suggests hope for future investigations.
Collapse
Affiliation(s)
- Wei Guo
- Department of Pharmacology, School of Pharmacy, Shanghai 201203, China
| | - Ze-yu Cheng
- Department of Pharmacology, School of Pharmacy, Shanghai 201203, China
| | - Yi-zhun Zhu
- Department of Pharmacology, School of Pharmacy, Shanghai 201203, China
- Institute of Biomedical Sciences, Fudan University, Shanghai 201203, China
- Department of Pharmacology, National University of Singapore, Singapore
| |
Collapse
|
57
|
The anti-apoptotic effect of hydrogen sulfide attenuates injuries to the medullary respiratory centers of neonatal rats subjected to in utero cigarette smoke exposure. Respir Physiol Neurobiol 2013; 188:29-38. [DOI: 10.1016/j.resp.2013.05.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/23/2013] [Accepted: 05/02/2013] [Indexed: 01/20/2023]
|
58
|
Shen Y, Guo W, Wang Z, Zhang Y, Zhong L, Zhu Y. Protective effects of hydrogen sulfide in hypoxic human umbilical vein endothelial cells: a possible mitochondria-dependent pathway. Int J Mol Sci 2013; 14:13093-108. [PMID: 23799362 PMCID: PMC3742176 DOI: 10.3390/ijms140713093] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 05/29/2013] [Accepted: 06/03/2013] [Indexed: 01/23/2023] Open
Abstract
The aim of the study was to investigate the protective effects of sodium hydrosulfide (NaHS), a H2S donor, against hypoxia-induced injury in human umbilical vein endothelial cells (HUVECs) and also to look into the possible mechanisms by which H2S exerts this protective effect. 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and scratch wound healing assay were chosen to measure the cell viability and migration-promoting effects. The fluorescent probe, DCFH-DA and 5,5′,6,6′-Tetrachloro-1,1′,3,3′-tetraethyl-imidacarbocyanine iodide (JC-1) were applied to detect the reactive oxygen species (ROS) level and mitochondrial membrane potential (ΔΨm). Furthermore, western blots were used to measure the expressions of the apoptosis-related proteins. Under hypoxic conditions, 300 μM and 600 μM of H2S could protect HUVECs against hypoxia-induced injury, as determined by MTT assay. Following the treatment of 60 μM NaHS for 18 h, scratch wound healing assays indicated that the scratch became much narrower than control group. After treatment with 60 μM, 120 μM, and 600 μM NaHS, and hypoxia for 30 min, flow cytometry demonstrated that the ROS concentrations decreased to 95.08% ± 5.52%, 73.14% ± 3.36%, and 73.51% ± 3.05%, respectively, compared with the control group. In addition, the JC-1 assay showed NaHS had a protective effect on mitochondria damage. Additionally, NaHS increased Bcl-2 expression and decreased the expression of Bax, Caspase-3 and Caspase-9 in a dose-dependent way. Our results suggest that H2S can protect endothelial cells and promote migration under hypoxic condition in HUVECs. These effects are partially associated with the preservation of mitochondrial function mediated by regulating the mitochondrial-dependent apoptotic pathway.
Collapse
Affiliation(s)
- Yaqi Shen
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China; E-Mails: (Y.S.); (W.G.); (Z.W.); (Y.Z.); (L.Z.)
| | - Wei Guo
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China; E-Mails: (Y.S.); (W.G.); (Z.W.); (Y.Z.); (L.Z.)
| | - Zhijun Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China; E-Mails: (Y.S.); (W.G.); (Z.W.); (Y.Z.); (L.Z.)
| | - Yuchen Zhang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China; E-Mails: (Y.S.); (W.G.); (Z.W.); (Y.Z.); (L.Z.)
| | - Liangjie Zhong
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China; E-Mails: (Y.S.); (W.G.); (Z.W.); (Y.Z.); (L.Z.)
| | - Yizhun Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China; E-Mails: (Y.S.); (W.G.); (Z.W.); (Y.Z.); (L.Z.)
- Institute of Biomedical Sciences, Fudan University, Shanghai 201203, China
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +862-151-980-018; Fax: +862-151-980-008
| |
Collapse
|
59
|
Huang CW, Lai MC, Cheng JT, Tsai JJ, Huang CC, Wu SN. Pregabalin attenuates excitotoxicity in diabetes. PLoS One 2013; 8:e65154. [PMID: 23785408 PMCID: PMC3681790 DOI: 10.1371/journal.pone.0065154] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2013] [Accepted: 04/22/2013] [Indexed: 12/15/2022] Open
Abstract
Diabetes can exacerbate seizures and worsen seizure-related brain damage. In the present study, we aimed to determine whether the standard antiepileptic drug pregabalin (PGB) protects against pilocarpine-induced seizures and excitotoxicity in diabetes. Adult male Sprague-Dawley rats were divided into either a streptozotocin (STZ)-induced diabetes group or a normal saline (NS) group. Both groups were further divided into subgroups that were treated intravenously with either PGB (15 mg/kg) or a vehicle; all groups were treated with subcutaneous pilocarpine (60 mg/kg) to induce seizures. To evaluate spontaneous recurrent seizures (SRS), PGB-pretreated rats were fed rat chow containing oral PGB (450 mg) for 28 consecutive days; vehicle-pretreated rats were fed regular chow. SRS frequency was monitored for 2 weeks from post-status epilepticus day 15. We evaluated both acute neuronal loss and chronic mossy fiber sprouting in the CA3 area. In addition, we performed patch clamp recordings to study evoked excitatory postsynaptic currents (eEPSCs) in hippocampal CA1 neurons for both vehicle-treated rats with SRS. Finally, we used an RNA interference knockdown method for Kir6.2 in a hippocampal cell line to evaluate PGB's effects in the presence of high-dose ATP. We found that compared to vehicle-treated rats, PGB-treated rats showed less severe acute seizure activity, reduced acute neuronal loss, and chronic mossy fiber sprouting. In the vehicle-treated STZ rats, eEPSC amplitude was significantly lower after PGB administration, but glibenclamide reversed this effect. The RNA interference study confirmed that PGB could counteract the ATP-sensitive potassium channel (KATP)-closing effect of high-dose ATP. By opening KATP, PGB protects against neuronal excitotoxicity, and is therefore a potential antiepileptogenic in diabetes. These findings might help develop a clinical algorithm for treating patients with epilepsy and comorbid metabolic disorders.
Collapse
Affiliation(s)
- Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- * E-mail: (CWH); (SNW)
| | - Ming-Chi Lai
- Department of Pediatrics, Chi-Mei Foundation Medical Center, Tainan City, Taiwan
| | - Juei-Tang Cheng
- Department of Pharmacology, National Cheng Kung University, Tainan, Taiwan
| | - Jing-Jane Tsai
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chao-Ching Huang
- Department of Pediatrics, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University, Tainan, Taiwan
- * E-mail: (CWH); (SNW)
| |
Collapse
|
60
|
Kotsyuba EP. Peculiarities of distribution of cystathionine β-synthase in the crustacean brain. J EVOL BIOCHEM PHYS+ 2013. [DOI: 10.1134/s002209301302014x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
61
|
Liu Y, Li Y, Yang W, Cao G. H2 S inhibits the activation of hepatic stellate cells and downregulates the expression of urotensin II. Hepatol Res 2013; 43:670-8. [PMID: 23131022 DOI: 10.1111/hepr.12002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Revised: 10/05/2012] [Accepted: 10/10/2012] [Indexed: 12/16/2022]
Abstract
AIM H2 S, a newly discovered signaling gasotransmitter, has been found involved in the pathogenesis of portal hypertension through the H2 S/CSE system. Studies also showed that urotensin II (UII), a recently discovered most potent vasoconstrictor, played an important role in cirrhotic portal hypertension. Therefore, studies were conducted to explore the relationship between H2 S and UII in cirrhosis. METHODS In the present study, the changes in the expression levels of cystathionine-γ-lyase (CSE), UII, urotensin II receptor (UT), collagen I, collagen III, tissue inhibitor of metalloproteinase-1 (TIMP-1) and α-smooth muscle actin (α-SMA) were determined by fluorescence quantitative polymerase chain reaction after exposure of hepatic stellate cells to H2 S. The influence of H2 S on UII was examined by western blotting, and the relationship between H2 S and UII was further confirmed by detection of cell proliferation and apoptosis. RESULTS Studies have shown that increase in H2 S concentration could reduce the expression of UII, UT, collagen I, collagen III, TIMP-1 and α-SMA without involvement of CSE. Moreover, the results of western blotting further proved that H2 S inhibited the expression of UII. The examination of cell proliferation by 5-ethynyl-2'-deoxyuridine assay suggests that H2 S significantly inhibited the proliferation of LX-2 cells and the proliferation-promoting effect of UII. Similarly, the examination of cell apoptosis revealed that H2 S could promote LX-2 cell apoptosis and inhibit the apoptosis-inhibiting effect of UII. CONCLUSION H2 S suppresses fibrosis by inhibiting the activation of hepatic stellate cells and reducing the expression of UII.
Collapse
Affiliation(s)
- Yang Liu
- Department of General Surgery, Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, China
| | | | | | | |
Collapse
|
62
|
Khademullah CS, Ferguson AV. Depolarizing actions of hydrogen sulfide on hypothalamic paraventricular nucleus neurons. PLoS One 2013; 8:e64495. [PMID: 23691233 PMCID: PMC3656899 DOI: 10.1371/journal.pone.0064495] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2013] [Accepted: 04/15/2013] [Indexed: 01/23/2023] Open
Abstract
Hydrogen sulfide (H2S) is a novel neurotransmitter that has been shown to influence cardiovascular functions as well and corticotrophin hormone (CRH) secretion. Since the paraventricular nucleus of the hypothalamus (PVN) is a central relay center for autonomic and endocrine functions, we sought to investigate the effects of H2S on the neuronal population of the PVN. Whole cell current clamp recordings were acquired from the PVN neurons and sodium hydrosulfide hydrate (NaHS) was bath applied at various concentrations (0.1, 1, 10, and 50 mM). NaHS (1, 10, and 50 mM) elicited a concentration-response relationship from the majority of recorded neurons, with almost exclusively depolarizing effects following administration. Cells responded and recovered from NaHS administration quickly and the effects were repeatable. Input differences from baseline and during the NaHS-induced depolarization uncovered a biphasic response, implicating both a potassium and non-selective cation conductance. The results from the neuronal population of the PVN shed light on the possible physiological role that H2S has in autonomic and endocrine function.
Collapse
Affiliation(s)
- C Sahara Khademullah
- Department of Biomedical and Molecular Science, Queen's University, Kingston, Ontario, Canada
| | | |
Collapse
|
63
|
Jia J, Xiao Y, Wang W, Qing L, Xu Y, Song H, Zhen X, Ao G, Alkayed NJ, Cheng J. Differential mechanisms underlying neuroprotection of hydrogen sulfide donors against oxidative stress. Neurochem Int 2013; 62:1072-8. [PMID: 23587562 DOI: 10.1016/j.neuint.2013.04.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2013] [Revised: 03/05/2013] [Accepted: 04/04/2013] [Indexed: 12/27/2022]
Abstract
This study investigated whether slow-releasing organic hydrogen sulfide donors act through the same mechanisms as those of inorganic donors to protect neurons from oxidative stress. By inducing oxidative stress in a neuronal cell line HT22 with glutamate, we investigated the protective mechanisms of the organic donors: ADT-OH [5-(4-hydroxyphenyl)-3H-1,2-dithiole-3-thione], the most widely used moiety for synthesizing slow-releasing hydrogen sulfide donors, and ADT, a methyl derivative of ADT-OH. The organic donors were more potent than the inorganic donor sodium hydrogensulfide (NaHS) in protecting HT22 cells against glutamate toxicity. Consistent with previous publications, NaHS partially restored glutamate-depleted glutathione (GSH) levels, protected HT22 from direct free radical damage induced by hydrogen peroxide (H2O2), and NaHS protection was abolished by a KATP channel blocker glibenclamide. However, neither ADT nor ADT-OH enhanced glutamate-depleted GSH levels or protected HT22 from H2O2-induced oxidative stress. Glibenclamide, which abolished NaHS neuroprotection against oxidative stress, did not block ADT and ADT-OH neuroprotection against glutamate-induced oxidative stress. Unexpectedly, we found that glutamate induced AMPK activation and that compound C, a well-established AMPK inhibitor, remarkably protected HT22 from glutamate-induced oxidative stress, suggesting that AMPK activation contributed to oxidative glutamate toxicity. Interestingly, all hydrogen sulfide donors, including NaHS, remarkably attenuated glutamate-induced AMPK activation. However, under oxidative glutamate toxicity, compound C only increased the viability of HT22 cells treated with NaHS, but did not further increase ADT and ADT-OH neuroprotection. Thus, suppressing AMPK activation likely contributed to ADT and ADT-OH neuroprotection. In conclusion, hydrogen sulfide donors acted through differential mechanisms to confer neuroprotection against oxidative toxicity and suppressing AMPK activation was a possible mechanism underlying neuroprotection of organic hydrogen sulfide donors against oxidative toxicity.
Collapse
Affiliation(s)
- Jia Jia
- Department of Pharmacology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
64
|
Zhou J, Wu PF, Wang F, Chen JG. Targeting gaseous molecules to protect against cerebral ischaemic injury: mechanisms and prospects. Clin Exp Pharmacol Physiol 2013; 39:566-76. [PMID: 22150768 DOI: 10.1111/j.1440-1681.2011.05654.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
1. Ischaemic brain injury is a leading cause of death and disability in many countries. However, the pathological mechanisms underlying ischaemic brain injury, including oxidative stress, calcium overload, excitotoxicity and neuronal apoptosis, are perplexing and this makes it difficult to find effective novel drugs for the treatment of the condition. 2. Recently, gaseous molecules such as nitric oxide (NO), carbon monoxide (CO), hydrogen sulphide (H(2)S) and hydrogen (H(2)) have attracted considerable interest because of their physiological and pathophysiological roles in various body systems. Emerging evidence indicates that gaseous molecules are involved in the pathological processes of ischaemic brain damage. 3. In the present review, we summarize evidence regarding the involvement of gaseous molecules in ischaemic brain injury and discuss the therapeutic potential of targeting gaseous molecules. 4. Collectively, the available data suggest that the application of these biological gas molecules and their pharmacological regulators may be a potential therapeutic approach for the treatment of ischaemic brain injury.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, #13 Hangkong Road, Wuhan, Hubei 430030, China
| | | | | | | |
Collapse
|
65
|
Dynamic Change of Hydrogen Sulfide After Traumatic Brain Injury and its Effect in Mice. Neurochem Res 2013; 38:714-25. [DOI: 10.1007/s11064-013-0969-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2012] [Revised: 12/14/2012] [Accepted: 01/08/2013] [Indexed: 01/20/2023]
|
66
|
Park SJ, Kim TH, Lee SH, Ryu HY, Hong KH, Jung JY, Hwang GH, Lee SH. Expression levels of endogenous hydrogen sulfide are altered in patients with allergic rhinitis. Laryngoscope 2013; 123:557-63. [PMID: 23303708 DOI: 10.1002/lary.23466] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2012] [Revised: 04/04/2012] [Accepted: 05/04/2012] [Indexed: 11/06/2022]
Abstract
OBJECTIVES/HYPOTHESIS Hydrogen sulfide (H(2) S), the third endogenous gaseous transmitter, may be a crucial mediator in airway hyper-responsiveness and airway inflammation, including asthma. To elucidate the role of H(2) S in allergic rhinitis, the present study was undertaken to determine the level of expression of H(2) S in healthy nasal mucosa and mild and moderate/severe persistent allergic nasal mucosa as well as peripheral blood obtained from each patient. The expression and distribution pattern of the H(2) S-synthesizing enzymes cystathione γ-lyase (CSE) and cystathione β-synthase (CBS) were investigated in healthy and allergic nasal mucosa. STUDY DESIGN Controlled, prospective study. METHODS The concentration of H(2) S in nasal mucosa and plasma was determined by zinc trap spectrophotometry. The expression levels and patterns of distribution of CSE and CBS mRNA and proteins were evaluated using real time polymerase chain reaction, Western blot, and immunohistochemistry. RESULTS The levels of expression of H(2) S in nasal mucosa and plasma were increased in patients with mild and moderate/severe persistent allergic rhinitis compared with healthy controls. CSE was localized in vascular endothelium and surrounding muscles, and submucosal glands, whereas CBS was exclusively distributed in the superficial epithelium and submucosal glands. Their expression levels were increased in mild and moderate/severe persistent allergic rhinitis. CONCLUSIONS The current findings indicate that, in parallel with increased expression levels of CSE and CBS, H(2) S is upregulated in nasal mucosa and plasma of allergic patients. Based on localization of CSE and CBS, H(2) S may play multiple functions in human nasal mucosa, contributing to the development of allergic symptoms such as rhinorrhea, sneezing, and nasal stuffiness.
Collapse
Affiliation(s)
- Se Jin Park
- Department of Otorhinolaryngology-Head and Neck Surgery, College of Medicine, Korea University, Seoul, South Korea
| | | | | | | | | | | | | | | |
Collapse
|
67
|
Liu YY, Nagpure BV, Wong PTH, Bian JS. Hydrogen sulfide protects SH-SY5Y neuronal cells against d-galactose induced cell injury by suppression of advanced glycation end products formation and oxidative stress. Neurochem Int 2012; 62:603-9. [PMID: 23274001 DOI: 10.1016/j.neuint.2012.12.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Revised: 11/29/2012] [Accepted: 12/18/2012] [Indexed: 10/27/2022]
Abstract
d-Galactose is widely used as an agent to cause aging effects in experimental animals. The present study aims to investigate the effects of hydrogen sulfide (H2S) in human neuroblastoma SH-SY5Y cells exposed to d-galactose. Cells were pretreated with NaHS, an H2S donor, and then exposed to d-galactose (25-400mM for 48h). We found that NaHS pretreatment significantly reversed the d-galactose-induced cell death and cellular senescence. MTT assay shows that NaHS significantly increased cell viability from 62.31±1.29% to 72.34±0.46% compared with d-galactose (200mM) treatment group. The underlying mechanism appeared to involve a reduction by NaHS in the formation of advanced glycation end products (AGEs), which are known to contribute to the progression of age-related diseases. In addition, NaHS decreased the elevation of reactive oxygen species from 151.17±2.07% to 124.8±2.89% and malondialdehyde from 1.72±0.07 to 1.10±0.08 (nmol/mg protein) in SH-SY5Y cells after d-galactose exposure. NaHS also stimulated activities of superoxide dismutase from 0.42±0.05 to 0.73±0.04 (U/mg protein) and glutathione peroxidase from 3.98±0.73 to 14.73±0.77 (nmol/min/mg protein) and upregulated the gene expression levels of copper transport protein ATOX1, glutathione synthetase (GSS) and thioredoxin reductase 1 (TXNRD1) while down-regulated aldehyde oxidase 1 (AOX1). In summary, our data indicate that H2S may have potentially anti-aging effects through the inhibition of AGEs formation and reduction of oxidative stress.
Collapse
Affiliation(s)
- Yan-Ying Liu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
| | | | | | | |
Collapse
|
68
|
Cui J, Liu L, Zou J, Qiao W, Liu H, Qi Y, Yan C. Protective effect of endogenous hydrogen sulfide against oxidative stress in gastric ischemia-reperfusion injury. Exp Ther Med 2012; 5:689-694. [PMID: 23403765 PMCID: PMC3570130 DOI: 10.3892/etm.2012.870] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 12/03/2012] [Indexed: 12/31/2022] Open
Abstract
Hydrogen sulfide (H2S) is a gaseous signaling molecule, which plays a critical role in a number of physiological and pathological progresses. In order to determine the effect of endogenous H2S on gastric ischemia-reperfusion (GI-R), we evaluated the gastric mucosal damage in rats intraperitoneally injected with DL-propargylglycine (PAG, 50 mg/kg/day) or L-cysteine (L-cys, 50 mg/kg/day) for 7 days before GI-R. GI-R injury was achieved by clamping the celiac artery for 30 min, followed by reperfusion for 60 min. Gastric mucosal damage was macroscopically assessed in the area of injury and deep damage was assessed by histopathological scoring. PAG increased the area of gastric mucosal injury and deep damage compared with that in untreated GI-R rats (P<0.05). While PAG decreased the H2S concentration and cystathionine γ-lyase (CSE) expression in the gastric mucosa, L-cys significantly attenuated the effects of GI-R. Western blot analysis revealed that the increases of malondialdehyde (MDA) and xanthine oxidase (XOD), and decreases of glutathione (GSH), superoxide dismutase (SOD) and the restriction of superoxide (O2−) production in the PAG group were inhibited by L-cys (P<0.05). Endogenous H2S has a protective effect against GI-R in rats by inhibiting oxygen free radical overproduction.
Collapse
Affiliation(s)
- Jie Cui
- Department of Physiology, Xuzhou Medical College, Xuzhou 221004, P.R. China
| | | | | | | | | | | | | |
Collapse
|
69
|
Iranon NN, Miller DL. Interactions between oxygen homeostasis, food availability, and hydrogen sulfide signaling. Front Genet 2012; 3:257. [PMID: 23233860 PMCID: PMC3516179 DOI: 10.3389/fgene.2012.00257] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2012] [Accepted: 11/04/2012] [Indexed: 12/19/2022] Open
Abstract
The ability to sense and respond to stressful conditions is essential to maintain organismal homeostasis. It has long been recognized that stress response factors that improve survival in changing conditions can also influence longevity. In this review, we discuss different strategies used by animals in response to decreased O(2) (hypoxia) to maintain O(2) homeostasis, and consider interactions between hypoxia responses, nutritional status, and H(2)S signaling. O(2) is an essential environmental nutrient for almost all metazoans as it plays a fundamental role in development and cellular metabolism. However, the physiological response(s) to hypoxia depend greatly on the amount of O(2) available. Animals must sense declining O(2) availability to coordinate fundamental metabolic and signaling pathways. It is not surprising that factors involved in the response to hypoxia are also involved in responding to other key environmental signals, particularly food availability. Recent studies in mammals have also shown that the small gaseous signaling molecule hydrogen sulfide (H(2)S) protects against cellular damage and death in hypoxia. These results suggest that H(2)S signaling also integrates with hypoxia response(s). Many of the signaling pathways that mediate the effects of hypoxia, food deprivation, and H(2)S signaling have also been implicated in the control of lifespan. Understanding how these pathways are coordinated therefore has the potential to reveal new cellular and organismal homeostatic mechanisms that contribute to longevity assurance in animals.
Collapse
Affiliation(s)
- Nicole N Iranon
- Department of Biochemistry, University of Washington School of Medicine Seattle, WA, USA ; Molecular and Cellular Biology Graduate Program, University of Washington School of Medicine Seattle, WA, USA
| | | |
Collapse
|
70
|
Abstract
Hydrogen sulfide (H(2)S) has been found to play an important role as a signal molecule in regulating cell survival. It appears paradoxical that, on one side, H(2)S acts as a physiological intercellular messenger to stimulate cell growth, and on the other side, it may display cytotoxic activity. This article summarizes the current body of evidence demonstrating the cytoprotective versus cytotoxic effects of H(2)S in mammalian cells and describes the janus-faced properties of this important gasotransmitter. This article will also provide a brief description of the current signaling mechanisms that have been demonstrated to be responsible for these different actions. The pharmacologic regulation of H(2)S production and the potential clinical significance of H(2)S are highlighted.
Collapse
Affiliation(s)
- Guangdong Yang
- The School of Kinesiology, Lakehead University, 955 Oliver Road, Thunder Bay, Ontario, P7B 5E1, Canada.
| |
Collapse
|
71
|
Esposito S, Pristerà A, Maresca G, Cavallaro S, Felsani A, Florenzano F, Manni L, Ciotti MT, Pollegioni L, Borsello T, Canu N. Contribution of serine racemase/d-serine pathway to neuronal apoptosis. Aging Cell 2012; 11:588-98. [PMID: 22507034 DOI: 10.1111/j.1474-9726.2012.00822.x] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Recent data indicate that age-related N-methyl-d-aspartate receptor (NMDAR) transmission impairment is correlated with the reduction in serine racemase (SR) expression and d-serine content. As apoptosis is associated with several diseases and conditions that generally occur with age, we investigated the modulation of SR/d-serine pathway during neuronal apoptosis and its impact on survival. We found that in cerebellar granule neurons (CGNs), undergoing apoptosis SR/d-serine pathway is crucially regulated. In the early phase of apoptosis, the expression of SR is reduced, both at the protein and RNA level through pathways, upstream of caspase activation, involving ubiquitin proteasome system (UPS) and c-Jun N-terminal kinases (JNKs). Forced expression of SR, together with treatment with NMDA and d-serine, blocks neuronal death, whereas pharmacological inhibition and Sh-RNA-mediated suppression of endogenous SR exacerbate neuronal death. In the late phase of apoptosis, the increased expression of SR contribute to the last, NMDAR-mediated, wave of cell death. These findings are relevant to our understanding of neuronal apoptosis and NMDAR activity regulation, raising further questions as to the role of SR/d-serine in those neuro-pathophysiological processes, such as aging and neurodegenerative diseases characterized by a convergence of apoptotic mechanisms and NMDAR dysfunction.
Collapse
Affiliation(s)
- Simona Esposito
- Istituto di Biologia Cellulare e Neurobiologia, CNR, Roma, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
72
|
Hydrogen sulfide prevents hypoxia-induced apoptosis via inhibition of an H2O2-activated calcium signaling pathway in mouse hippocampal neurons. Biochem Biophys Res Commun 2012; 425:473-7. [PMID: 22846576 DOI: 10.1016/j.bbrc.2012.07.131] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Accepted: 07/21/2012] [Indexed: 11/22/2022]
Abstract
Hydrogen sulfide (H(2)S), an endogenous gaseous mediator, has been shown to exert protective effects against damage to different organs in the human body caused by various stimuli. However, the potential effects of H(2)S on hypoxia-induced neuronal apoptosis and its mechanisms remain unclear. Here, we exposed mouse hippocampal neurons to hypoxic conditions (2% O(2), 5% CO(2) and 93% N(2) at 37 °C) to establish a hypoxic cell model. We found that 4-h hypoxia treatment significantly increased intracellular reactive oxygen species (ROS) levels, and pretreatment with NaHS (a source of H(2)S) for 30 min suppressed hypoxia-induced intracellular ROS elevation. The hypoxia treatment significantly increased cytosolic calcium ([Ca(2+)](i)), and pretreatment with NaHS prevented the increase in [Ca(2+)](i). Additionally, polyethylene glycol (PEG)-catalase (a H(2)O(2) scavenger) but not PEG-SOD (an O(2)(-) scavenger) conferred an inhibitory effect similar to H(2)S on the hypoxia-induced increase in [Ca(2+)](i). Furthermore, we found that pretreatment with NaHS could significantly inhibit hypoxia-induced neuronal apoptosis, which was also inhibited by PEG-catalase or the inositol 1,4,5-triphosphate (IP(3)) receptor blocker xestospongin C. Taken together, these findings suggest that H(2)S inhibits hypoxia-induced apoptosis through inhibition of a ROS (mainly H(2)O(2))-activated Ca(2+) signaling pathway in mouse hippocampal neurons.
Collapse
|
73
|
Marutani E, Kosugi S, Tokuda K, Khatri A, Nguyen R, Atochin DN, Kida K, Van Leyen K, Arai K, Ichinose F. A novel hydrogen sulfide-releasing N-methyl-D-aspartate receptor antagonist prevents ischemic neuronal death. J Biol Chem 2012; 287:32124-35. [PMID: 22815476 DOI: 10.1074/jbc.m112.374124] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Physiological levels of H(2)S exert neuroprotective effects, whereas high concentrations of H(2)S may cause neurotoxicity in part via activation of NMDAR. To characterize the neuroprotective effects of combination of exogenous H(2)S and NMDAR antagonism, we synthesized a novel H(2)S-releasing NMDAR antagonist N-((1r,3R,5S,7r)-3,5-dimethyladamantan-1-yl)-4-(3-thioxo-3H-1,2-dithiol-4-yl)-benzamide (S-memantine) and examined its effects in vitro and in vivo. S-memantine was synthesized by chemically combining a slow releasing H(2)S donor 4-(3-thioxo-3H-1,2-dithiol-4-yl)-benzoic acid (ACS48) with a NMDAR antagonist memantine. S-memantine increased intracellular sulfide levels in human neuroblastoma cells (SH-SY5Y) 10-fold as high as that was achieved by ACS48. Incubation with S-memantine after reoxygenation following oxygen and glucose deprivation (OGD) protected SH-SY5Y cells and murine primary cortical neurons more markedly than did ACS48 or memantine. Glutamate-induced intracellular calcium accumulation in primary cortical neurons were aggravated by sodium sulfide (Na(2)S) or ACS48, but suppressed by memantine and S-memantine. S-memantine prevented glutamate-induced glutathione depletion in SH-SY5Y cells more markedly than did Na(2)S or ACS48. Administration of S-memantine after global cerebral ischemia and reperfusion more robustly decreased cerebral infarct volume and improved survival and neurological function of mice than did ACS48 or memantine. These results suggest that an H(2)S-releasing NMDAR antagonist derivative S-memantine prevents ischemic neuronal death, providing a novel therapeutic strategy for ischemic brain injury.
Collapse
Affiliation(s)
- Eizo Marutani
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Charlestown, Massachusetts 02129, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
74
|
Abstract
SIGNIFICANCE Evidence of the ability of the gasotransmitter hydrogen sulfide (H(2)S) to serve as a regulator of many physiological functions, including control of blood pressure, regulation of cardiac function, protection of neurons, and cardiomyocytes against apoptosis, and in pain sensation is accumulating. However, the mechanisms accounting for its many actions are not yet well understood. RECENT ADVANCES Following the pioneering studies of the regulation of N-methyl-d-aspartate receptors and ATP-sensitive K(+) channels by H(2)S, data continue to emerge indicating that H(2)S modulates other ion channel types. This article reviews the numerous, yet diverse, types of ion channels now reported to be regulated by H(2)S. CRITICAL ISSUES Currently, a critical issue within this field is to determine the mechanisms by which H(2)S regulates ion channels, as well as other target proteins. Mechanisms to account for regulation include direct channel protein sulfhydration, channel redox modulation, effects mediated by interactions with other gasotransmitters (carbon monoxide and nitric oxide), and indirect effects, such as modulation of channel-regulating kinases. Through such modulation of ion channels, novel roles for H(2)S are emerging as important factors in both physiological and pathological processes. FUTURE DIRECTIONS Increasing current awareness and understanding of the roles and mechanisms of action of ion channel regulation by H(2)S will open opportunities for therapeutic intervention with clear clinical benefits, and inform future therapies. In addition, more sensitive methods for detecting relevant physiological concentrations of H(2)S will allow for clarification of specific ion channel regulation with reference to physiological or pathophysiological settings.
Collapse
Affiliation(s)
- Chris Peers
- Division of Cardiovascular and Neuronal Remodelling, Faculty of Medicine and Health, Leeds Institute for Genetics Health and Therapeutics, University of Leeds, Leeds, United Kingdom.
| | | | | | | | | |
Collapse
|
75
|
Abstract
SIGNIFICANCE An abundance of experimental evidence suggests that hydrogen sulfide (H(2)S) plays a prominent role in physiology and pathophysiology. Many targets exist for H(2)S therapy. The molecular targets of H(2)S include proteins, enzymes, transcription factors, and membrane ion channels. RECENT ADVANCES Novel H(2)S precursors are being synthesized and discovered that are capable of releasing H(2)S in a slow and sustained manner. This presents a novel and advantageous approach to H(2)S therapy for treatment of chronic conditions associated with a decline in endogenous H(2)S, such as diabetes and cardiovascular disease. CRITICAL ISSUES While H(2)S is cytoprotective at physiological concentrations, it is not universally cytoprotective, as it appears to have pro-apoptotic actions in cancer cells and is well known to be toxic at supraphysiological concentrations. Many of the pleiotropic effects of H(2)S on health are associated with the inhibition of inflammation and upregulation of prosurvival pathways. The powerful anti-inflammatory, cytoprotective, immunomodulating, and trophic effects of H(2)S on the vast majority of normal cells seem to be mediated mainly by its actions as an extremely versatile direct and indirect antioxidant and free radical scavenger. While the overall effects of H(2)S on transformed (i.e., malignant) cells can be characterized as pro-oxidant and pro-apoptotic, they contrast sharply with the cytoprotective effects on most normal cells. FUTURE DIRECTIONS H(2)S has become a molecule of great interest, and several slow-releasing H(2)S prodrugs are currently under development. We believe that additional agents regulating H(2)S bioavailability will be developed during the next 10 years.
Collapse
Affiliation(s)
- Benjamin Lee Predmore
- Department of Surgery-Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | |
Collapse
|
76
|
Xie L, Tiong CX, Bian JS. Hydrogen sulfide protects SH-SY5Y cells against 6-hydroxydopamine-induced endoplasmic reticulum stress. Am J Physiol Cell Physiol 2012; 303:C81-91. [PMID: 22555844 DOI: 10.1152/ajpcell.00281.2011] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Endoplasmic reticulum (ER) stress has been implicated in several neurodegenerative diseases, including Parkinson's disease. The present study attempted to investigate the effect of hydrogen sulfide (H(2)S) on 6-hydroxydopamine (6-OHDA)-induced ER stress in SH-SY5Y cells. We found in the present study that exogenous application of sodium hydrosulfide (NaHS; an H(2)S donor, 100 μM) significantly attenuated 6-OHDA (50 μM)-induced cell death. NaHS also reversed the upregulation of cleaved poly(ADP-ribose) polymerase and caspase 9 in 6-OHDA-treated cells. Consistent with its cytoprotective effects, NaHS markedly reduced 6-OHDA induced-ER stress responses, including the upregulated levels of eukaryotic initiation factor-2α phosphorylation, glucose-regulated protein 78, and C/EBP homologous protein expression. The protective effect of H(2)S on ER stress was attenuated by blockade of Akt activity with an Akt inhibitor or inhibition of heat shock protein (Hsp)90 with geldanamycin but not by suppression of ERK1/2 with PD-98059. Blockade of Akt also significantly decreased the protein abundance of Hsp90 in SH-SY5Y cells. Moreover, overexpression of cystathionine β-synthase (a main H(2)S-synthesizing enzyme in the brain) elevated the Hsp90 protein level and suppressed 6-OHDA-induced ER stress. In conclusion, the protective effect of H(2)S against 6-OHDA-induced ER stress injury in SH-SY5Y cells involves the Akt-Hsp90 pathway.
Collapse
Affiliation(s)
- Li Xie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | |
Collapse
|
77
|
Kotsyuba EP. NO- and H2S brain systems of the Japanese shore crab Hemigrapsus sanguineus under conditions of anoxia. BIOL BULL+ 2012. [DOI: 10.1134/s1062359012030041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
78
|
Xie X, Sun A, Zhu W, Huang Z, Hu X, Jia J, Zou Y, Ge J. Transplantation of mesenchymal stem cells preconditioned with hydrogen sulfide enhances repair of myocardial infarction in rats. TOHOKU J EXP MED 2012; 226:29-36. [PMID: 22186034 DOI: 10.1620/tjem.226.29] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Stem cell transplantation has become a promising therapeutic approach for the treatment of myocardial infarction (MI). However, the poor survival of the donor cells after transplantation has restricted its therapeutic efficacy. Hydrogen sulfide (H(2)S), one gaseous signaling molecule, has been applied to inhibit cell apoptosis and promote cell survival. In the present study, we therefore examined the effects of H(2)S on the survival of mesenchymal stem cells (MSCs). MSCs were isolated from the femur of male Sprague-Dawley rats (about 4 weeks old, 100 g). Preconditioning MSCs with 200 µmol/L NaHS (as the donor of H(2)S) for 30 min decreased the hypoxia-induced cell apoptosis in vitro. The mechanisms contributing to the beneficial effects of H(2)S on MSCs were associated with increased levels of phosphorylated Akt (pAkt), phosphorylated Erk1/2 (pErk1/2) and phosphorylated glycogen synthase kinase-3β (pGSK-3β) in MSCs. Subsequently, MSCs (1 × 10(6)), MSCs preconditioned with H(2)S (1 × 10(6)), or phosphate buffered saline (PBS) were injected into rat hearts immediately after MI (the ligation of the left anterior descending of coronary artery). Real-time PCR for the Sry gene, located on the Y chromosome, indicated that preconditioning with H(2)S improved the survival rate of the transplanted MSCs in infarcted myocardium 4 days after MI, compared with the untreated MSCs. Furthermore, transplantation of the H(2)S-pretreated MSCs reduced the infarct size and increased left ventricular (LV) function, as judged by transthoracic echocardiography. In conclusion, H(2)S preconditioning effectively promotes MSCs survival under ischemic injury and helps cardiac repair after MI, which has great clinical significance.
Collapse
Affiliation(s)
- Xinxing Xie
- Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, PR China
| | | | | | | | | | | | | | | |
Collapse
|
79
|
Nagel S, Papadakis M, Pfleger K, Grond-Ginsbach C, Buchan AM, Wagner S. Microarray analysis of the global gene expression profile following hypothermia and transient focal cerebral ischemia. Neuroscience 2012; 208:109-22. [PMID: 22366221 DOI: 10.1016/j.neuroscience.2012.01.048] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2011] [Revised: 01/26/2012] [Accepted: 01/30/2012] [Indexed: 01/09/2023]
Abstract
BACKGROUND Hypothermia is one of the most robust experimental neuroprotective interventions against cerebral ischemia. Identification of molecular pathways and gene networks together with single genes or gene families that are significantly associated with neuroprotection might help unravel the mechanisms of therapeutic hypothermia. MATERIAL AND METHODS We performed a microarray analysis of ischemic rat brains that underwent 90 min of middle cerebral artery occlusion (MCAO) and 48 h of reperfusion. Hypothermia was induced for 4 h, starting 1 h after MCAO in male Wistar rats. At 48 h, magnetic resonance imaging (MRI) was performed for infarct volumetry, and functional outcome was determined by a neuroscore. The brain gene expression profile of sham (S), ischemia (I), and ischemia plus hypothermia (HI) treatment were compared by analyzing changes of individual genes, pathways, and networks. Real-time reverse-transcribed polymerase chain reaction (RT-PCR) was performed on selected genes to validate the data. RESULTS Rats treated with HI had significantly reduced infarct volumes and improved neuroscores at 48 h compared with I. Of 4067 genes present on the array chip, HI compared with I upregulated 50 (1.23%) genes and downregulated 103 (3.20%) genes equal or greater than twofold. New genes potentially mediating neuroprotection by hypothermia were HNRNPAB, HIG-1, and JAK3. On the pathway level, HI globally suppressed the ischemia-driven gene response. Twelve gene networks were identified to be significantly altered by HI compared with I. The most significantly altered network contained genes participating in apoptosis suppression. CONCLUSIONS Our data suggest that although hypothermia at the pathway level restored gene expression to sham levels, it selectively regulated the expression of several genes implicated in protein synthesis and folding, calcium homeostasis, cellular and synaptic integrity, inflammation, cell death, and apoptosis.
Collapse
Affiliation(s)
- S Nagel
- Department of Neurology, University of Heidelberg, Heidelberg, Germany.
| | | | | | | | | | | |
Collapse
|
80
|
Osborne NN, Ji D, Majid ASA, Del Soldata P, Sparatore A. Glutamate oxidative injury to RGC-5 cells in culture is necrostatin sensitive and blunted by a hydrogen sulfide (H2S)-releasing derivative of aspirin (ACS14). Neurochem Int 2012; 60:365-78. [PMID: 22306773 DOI: 10.1016/j.neuint.2012.01.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2011] [Revised: 01/03/2012] [Accepted: 01/09/2012] [Indexed: 01/04/2023]
Abstract
Oxidative stress to RGC-5 cells in culture was delivered by exposure to a combination of glutamate (Glu) and buthionine-S,R-sulfoximine (BSO). The effect of the insult on cell survival was quantified by the resazurin-reduction and a dead/live assays. Moreover, breakdown of DNA, the localisation of phosphatidylserine and reactive radical species (ROS) and its quantification were determined. In addition, various proteins and mRNAs were studied using Western blot, real time PCR and immunocytochemistry. ACS14, its sulfurated moiety ACS1 and aspirin were tested for their ability to blunt the negative effects of Glu/BSO on RGC-5 cells. In addition assays were carried out to see whether any of these substances influenced glutathione (GSH). Glu/BSO dose-dependently kills RGC-5 cells by a mechanism that involves an elevation of ROS accompanied by a breakdown of DNA, expression of phosphatidylserine and the activation of p38 MAPK. The process is unaffected by the pan caspase inhibitor z-VAD-fmk, does not involve the activation of apoptosis inducing factor (AIF) but is sensitive to active necrostatin-1. In cell viability studies (resazurin-reduction assay), ACS1 and ACS14 equally counteracted the negative effects of 5mM Glu/BSO to RGC-5 cells but aspirin was only effective with a milder oxidative stress (1 mM Glu/BSO). In all other assays ACS14 was very much more effective than aspirin at counteracting the influence of 5mM Glu/BSO. Moreover, ACS14 and ACS1 directly stimulated GSH while aspirin was ineffective. In addition the neuroprotecive effect of ACS14 was specifically blunted by the non-specific potassium channel blocker glibenclamide. Also the up-regulation of Bcl-2, HO-1 and XIAP induced by 5mM Glu/BSO were all attenuated to a greater extent by ACS14 (20 μM) than aspirin (20 μM). These data show that ACS14 is a very effective neuroprotectant when compared with aspirin. ACS14 maintains its aspirin characteristics and has the ability to release H(2)S. The combined multiple actions of aspirin and H(2)S in the form of ACS14 is worthy to consider for possible use in the treatment of glaucoma.
Collapse
Affiliation(s)
- Neville N Osborne
- Nuffield Laboratory of Ophthalmology, University of Oxford, John Radcliffe Hospital, Oxford, UK.
| | | | | | | | | |
Collapse
|
81
|
Pupo E, Pla AF, Avanzato D, Moccia F, Cruz JEA, Tanzi F, Merlino A, Mancardi D, Munaron L. Hydrogen sulfide promotes calcium signals and migration in tumor-derived endothelial cells. Free Radic Biol Med 2011; 51:1765-73. [PMID: 21875664 DOI: 10.1016/j.freeradbiomed.2011.08.007] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2011] [Revised: 08/08/2011] [Accepted: 08/10/2011] [Indexed: 12/15/2022]
Abstract
Hydrogen sulfide (H(2)S) is a gasotransmitter that plays several roles in various tissues, including the cardiovascular system. Because it has been recently proposed to act as a mediator of angiogenesis progression, here we investigate the effects of H(2)S in a well-established model of tumor angiogenesis: endothelial cells obtained from human breast carcinoma (B-TECs). Ca(2+) imaging and patch-clamp experiments reveal that acute perfusion with NaHS, a widely employed H(2)S donor, activates cytosolic calcium (Ca(c)) increase, as well as potassium and nonselective cationic currents, in B-TECs. Stimulation with NaHS in the same concentration range (1 nM-200 μM) evoked Ca(c) signals also in "normal" human microvascular endothelial cells (HMVECs), but the amplitude was significantly lower. Moreover, although NaHS failed to promote either migration or proliferation on HMVECs, B-TEC migration was enhanced at low-micromolar NaHS concentrations (1-10 μM). Remarkably H(2)S mediates tumor proangiogenic signaling triggered by vascular endothelial growth factor (VEGF). B-TECs pretreated with dl-propargylglycine (5mM, 30 min), an inhibitor of the H(2)S-producing enzyme cystathionine γ-lyase, showed drastically reduced migration and Ca(c) signals induced by VEGF (20 ng/ml). We conclude that H(2)S plays a role in proangiogenic signaling of tumor-derived but not normal human ECs. Furthermore the ability of this gasotransmitter to interfere with B-TEC responsiveness to VEGF suggests that it could be an interesting target for antiangiogenic strategies in tumor treatment.
Collapse
Affiliation(s)
- Emanuela Pupo
- Department of Animal and Human Biology, University of Torino, 10123 Torino, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Tang XQ, Zhuang YY, Fan LL, Fang HR, Zhou CF, Zhang P, Hu B. Involvement of KATP/PI3K/AKT/Bcl-2 Pathway in Hydrogen Sulfide-induced Neuroprotection Against the Toxicity of 1-methy-4-phenylpyridinium Ion. J Mol Neurosci 2011; 46:442-9. [DOI: 10.1007/s12031-011-9608-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2011] [Accepted: 07/20/2011] [Indexed: 12/21/2022]
|
83
|
Abstract
Hydrogen sulfide (H(2)S) was known to be a toxic gas and an environmental hazard for many decades. However, it is now recognized that H(2)S may serve as a gaseous mediator that is endogenously produced to influence biological functions in mammalian. Together with nitric oxide and carbon monoxide, it forms the group of mediators that has been termed the "gasotransmitters." The past decade has seen an exponential growth of scientific interest in the physiological and pathological significance of H(2)S especially with respect to its role in the central nervous system and the cardiovascular system. In the central nervous system, H(2)S facilitates long-term potentiation and regulates intracellular calcium concentration and pH level in brain cells. Intriguingly, H(2)S produces antioxidant, anti-inflammatory, and antiapoptotic effects that may have relevance to neurodegenerative disorders. Abnormal generation and metabolism of H(2)S have been reported in the pathogenesis of ischemic stroke, Alzheimer's disease, Parkinson's disease, and recurrent febrile seizure. Exogenously applied H(2)S is demonstrated to have value for the treatment of febrile seizure and Parkinson's disease. This article presents an overview of current knowledge of H(2)S in relation to brain functions, with a special emphasis on its neuroprotective effects and the underlying cellular and molecular mechanisms.
Collapse
Affiliation(s)
- Li-Fang Hu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | | | | |
Collapse
|
84
|
Li Z, Wang Y, Xie Y, Yang Z, Zhang T. Protective Effects of Exogenous Hydrogen Sulfide on Neurons of Hippocampus in a Rat Model of Brain Ischemia. Neurochem Res 2011; 36:1840-9. [DOI: 10.1007/s11064-011-0502-6] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/09/2011] [Indexed: 12/20/2022]
|
85
|
Xu ZS, Wang XY, Xiao DM, Hu LF, Lu M, Wu ZY, Bian JS. Hydrogen sulfide protects MC3T3-E1 osteoblastic cells against H2O2-induced oxidative damage-implications for the treatment of osteoporosis. Free Radic Biol Med 2011; 50:1314-23. [PMID: 21354302 DOI: 10.1016/j.freeradbiomed.2011.02.016] [Citation(s) in RCA: 140] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2010] [Revised: 02/15/2011] [Accepted: 02/16/2011] [Indexed: 01/16/2023]
Abstract
Osteoporosis is a bone disease that leads to an increased risk of fracture. Oxidative damage is an important contributor to the morphological and functional changes in the development of osteoporosis. We found in this study that hydrogen sulfide (H2S), a novel endogenous gaseous mediator, protected MC3T3-E1 osteoblastic cells against hydrogen peroxide (H2O2)-induced oxidative injury. NaHS, an H2S donor, increased cell viability and reduced cell apoptosis caused by H2O2. NaHS also stimulated osteoblast proliferation by enhancing both transcription and activity of alkaline phosphatase in MC3T3-E1 osteoblastic cells. Moreover, treatment with NaHS stimulated the transcriptional level of osteocalcin, the main bone matrix protein, and the protein expression of collagen, a major constituent of bone tissue. The above effects were mediated by the antioxidant effect of H2S. NaHS reversed the reduced superoxide dismutase activity, decreased reactive oxygen species production, and suppressed NADPH oxidase activity in H2O2-treated osteoblasts. In addition, NaHS treatment also produced anti-inflammatory effects via inhibition of the production of nitric oxide and TNF-α, suggesting an anti-inflammatory effect of H2S. Cell viability and Western blotting analysis demonstrated that the protective effects of H2S were mediated by p38 and ERK1/2 MAPKs. In conclusion, H2S protects osteoblastic cells against oxidative stress-induced cell injury and suppression of proliferation and differentiation via a MAPK (p38 and ERK1/2)-dependent mechanism. Our findings suggest that H2S may have a potentially therapeutic value for osteoporosis.
Collapse
Affiliation(s)
- Zhong-Shi Xu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597 Singapore
| | | | | | | | | | | | | |
Collapse
|
86
|
Olson KR. The therapeutic potential of hydrogen sulfide: separating hype from hope. Am J Physiol Regul Integr Comp Physiol 2011; 301:R297-312. [PMID: 21543637 DOI: 10.1152/ajpregu.00045.2011] [Citation(s) in RCA: 134] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Hydrogen sulfide (H(2)S) has become the hot new signaling molecule that seemingly affects all organ systems and biological processes in which it has been investigated. It has also been shown to have both proinflammatory and anti-inflammatory actions and proapoptotic and anti-apoptotic effects and has even been reported to induce a hypometabolic state (suspended animation) in a few vertebrates. The exuberance over potential clinical applications of natural and synthetic H(2)S-"donating" compounds is understandable and a number of these function-targeted drugs have been developed and show clinical promise. However, the concentration of H(2)S in tissues and blood, as well as the intrinsic factors that affect these levels, has not been resolved, and it is imperative to address these points to distinguish between the physiological, pharmacological, and toxicological effects of this molecule. This review will provide an overview of H(2)S metabolism, a summary of many of its reported "physiological" actions, and it will discuss the recent development of a number of H(2)S-donating drugs that show clinical potential. It will also examine some of the misconceptions of H(2)S chemistry that have appeared in the literature and attempt to realign the definition of "physiological" H(2)S concentrations upon which much of this exuberance has been established.
Collapse
Affiliation(s)
- Kenneth R Olson
- Indiana University School of Medicine-South Bend, South Bend, Indiana 46617, USA.
| |
Collapse
|
87
|
Kulkarni M, Njie-Mbye YF, Okpobiri I, Zhao M, Opere CA, Ohia SE. Endogenous production of hydrogen sulfide in isolated bovine eye. Neurochem Res 2011; 36:1540-5. [PMID: 21533862 DOI: 10.1007/s11064-011-0482-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2011] [Indexed: 11/29/2022]
Abstract
Hydrogen sulfide (H(2)S) is a novel gasotransmitter with physiological and pathological functions in vascular homeostasis, cardiovascular system and central nervous system. In the present study, we determined the endogenous levels of H(2)S in various tissues of the bovine eye. We also examined the basal levels of H(2)S in response to donors (sodium hydrosulfide, NaHS and sodium sulfide, Na(2)S), substrate (L: -cysteine), inhibitors (propargylglycine, PAG and aminooxyacetic acid, AOA) and activator (S-adenosyl-L: -methionine, SAM) of this gas in the bovine retina. H(2)S was measured using a well established spectrophotometric method. The highest concentration of endogenous H(2)S was detected in cornea (19 ± 2.85 nmoles/mg protein, n = 6) and retina (17 ± 2.1 nmoles/mg protein, n = 6). Interestingly, H(2)S was not present in vitreous humor. The inhibitors of CSE and CBS; PAG (1 mM) and AOA (1 mM), significantly attenuated the production of H(2)S in the bovine retina by 56.8 and 42%, respectively. On the other hand the activator of CBS; SAM (100 μM), H(2)S donors; NaHS (1 μM) and Na(2)S (100 μM), significantly increased endogenous levels of H(2)S in bovine retina. L: -cysteine (10-300 μM) produced a significant (P < 0.05) concentration-dependent increase in H(2)S levels reaching a maximal at 300 μM. We conclude that H(2)S is endogenously produced in various tissues of the isolated bovine eye. Moreover, endogenous levels of H(2)S are enhanced in the presence of substrate (L: -cysteine), an activator of CBS (SAM) and H(2)S donors but are blocked by inhibitors of enzymes that synthesize this gas in neural retina.
Collapse
Affiliation(s)
- Madhura Kulkarni
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, Texas Southern University, Houston, TX 77004, USA
| | | | | | | | | | | |
Collapse
|
88
|
Zhao ZZ, Wang Z, Li GH, Wang R, Tan JM, Cao X, Suo R, Jiang ZS. Hydrogen sulfide inhibits macrophage-derived foam cell formation. Exp Biol Med (Maywood) 2011; 236:169-76. [PMID: 21321313 DOI: 10.1258/ebm.2010.010308] [Citation(s) in RCA: 86] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Recent evidence indicates that hydrogen sulfide (H(2)S) exerts an antiatherogenic effect, but the mechanism is unclear. Formation of macrophage-derived foam cells is a crucial event in the development of atherosclerosis. Thus, we explore the effect of H(2)S on the formation of macrophage-derived foam cells. Incubation of monocyte-derived macrophages with oxidized LDL (oxLDL) alone caused significant increases both in intracellular lipids revealed by Oil-red O staining and in intracellular total cholesterol (TC) and esterified cholesterol (EC) concentrations assessed by high-performance liquid chromatography. Sodium hydrosulfide (NaHS, an H(2)S donor) remarkably abrogated oxLDL-induced intracellular lipid accumulation, and attenuated TC and EC concentrations and EC/TC ratio, whereas dl-propargylglycine (PPG) (a H(2)S-generating enzyme cystathionine gamma lyase inhibitor) exacerbated lipid accumulation and augmented TC and EC concentrations and EC/TC ratio. Incubation of 1,1'-dioctadecyl-3,3,3',3'-tetramethylindocarbocyanine perchlorate (DiI)-oxLDL led to lipoprotein binding and uptake of macrophages, which was blunted by NaHS, but enhanced by PPG. Furthermore, OxLDL markedly induced CD36, scavenger receptor A (SR-A) and acyl-coenzyme A:cholesterol acyltransferase-1 (ACAT-1) expressions in macrophages, which was suppressed by NaHS (50-200 μmol/L). Finally, the down-regulations of TC and EC concentrations as well as CD36 and ACAT-1 expressions by NaHS were suppressed by glibenclamide, a K(ATP) channel blocker, but facilitated by PD98059, an extracellular signal-regulated kinases 1 and 2 (ERK1/2) inhibitor. These results suggested that H(2)S inhibits foam cell formation by down-regulating CD36, SR-A and ACAT1 expressions via the K(ATP)/ERK1/2 pathway in human monocyte-derived macrophages.
Collapse
Affiliation(s)
- Zhan-Zhi Zhao
- Institute of Cardiovascular Disease and Key Laboratory for Arteriosclerology of Hunan Province, University of South China, Hengyang City, Hunan 421001, P. R. China
| | | | | | | | | | | | | | | |
Collapse
|
89
|
Tegeder I, Scheving R, Wittig I, Geisslinger G. SNO-ing at the nociceptive synapse? Pharmacol Rev 2011; 63:366-89. [PMID: 21436345 DOI: 10.1124/pr.110.004200] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Nitric oxide is generally considered a pronociceptive retrograde transmitter that, by activation of soluble guanylyl cyclase-mediated cGMP production and activation of cGMP-dependent protein kinase, drives nociceptive hypersensitivity. The duality of its functions, however, is increasingly recognized. This review summarizes nitric-oxide-mediated direct S-nitrosylation of target proteins that may modify nociceptive signaling, including glutamate receptors and G-protein-coupled receptors, transient receptor potential channels, voltage-gated channels, proinflammatory enzymes, transcription factors, and redoxins. S-Nitrosylation events require close proximity of nitric oxide production and target proteins and a permissive redox state in the vicinity. Despite the diversity of potential targets and effects, three major schemes arise that may affect nociceptive signaling: 1) S-Nitrosylation-mediated changes of ion channel gating properties, 2) modulation of membrane fusion and fission, and thereby receptor and channel membrane insertion, and 3) modulation of ubiquitination, and thereby protein degradation or transcriptional activity. In addition, S-Nitrosylation may alter the production of nitric oxide itself.
Collapse
Affiliation(s)
- Irmgard Tegeder
- Institut für Klinische Pharmakologie, Klinikum der Goethe-Universität Frankfurt, Theodor Stern Kai 7, Haus 74; 60590 Frankfurt am Main, Germany.
| | | | | | | |
Collapse
|
90
|
Meng JL, Mei WY, Dong YF, Wang JH, Zhao CM, Lan AP, Yang CT, Chen PX, Feng JQ, Hu CH. Heat shock protein 90 mediates cytoprotection by H2S against chemical hypoxia-induced injury in PC12 cells. Clin Exp Pharmacol Physiol 2010; 38:42-9. [DOI: 10.1111/j.1440-1681.2010.05462.x] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
91
|
Ren C, Du A, Li D, Sui J, Mayhan WG, Zhao H. Dynamic change of hydrogen sulfide during global cerebral ischemia–reperfusion and its effect in rats. Brain Res 2010; 1345:197-205. [DOI: 10.1016/j.brainres.2010.05.017] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2010] [Revised: 05/03/2010] [Accepted: 05/04/2010] [Indexed: 11/28/2022]
|