51
|
Bansal Y, Silakari O. Multifunctional compounds: smart molecules for multifactorial diseases. Eur J Med Chem 2014; 76:31-42. [PMID: 24565571 DOI: 10.1016/j.ejmech.2014.01.060] [Citation(s) in RCA: 146] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2012] [Accepted: 01/28/2014] [Indexed: 01/12/2023]
Abstract
Multifunctional compounds (MFCs) are designed broadly as hybrid or conjugated drugs or as chimeric drugs from two or more pharmacophores/drugs having specific pharmacological activities. These are capable of eliciting multiple pharmacological actions and have emerged as magic bullets in treatment of multifactorial diseases. Many research articles disclosing the development of such compounds for treatment of multifactorial diseases are published during last 7 years. Some successful MFC candidates for multifactorial CNS disorders include ziprasidone, duloxetine, ladostigil and M-30 whereas sunitinib, lapatinib and synthetic oleandane triterpinoids are the successful MFC candidates for various cancers. Many more compounds derived from berberine, tacrine, artemisnin, quinine, NSAIDs, pralidoxine, donepezil, rivastigmine, curcumin and various antioxidants are under investigations for exploration of their multifunctional potential. In general, MFCs possess the advantages of reduced molecularity, no drug-drug interactions and improved pharmacokinetics and pharmacodynamics. A MFC derived from two or more different pharmacophores exerts its activities by interacting with respective receptors of its constituent pharmacophores. It may also exhibit additional binding interactions with the receptor sites that may be responsible for significantly improved or additional activities. The present review discusses various MFCs developed for specific class of disorders with an aim to provide an insight into the strategies in medicinal chemistry for development of such compounds.
Collapse
Affiliation(s)
- Yogita Bansal
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India
| | - Om Silakari
- Molecular Modelling Lab (MML), Department of Pharmaceutical Sciences and Drug Research, Punjabi University, Patiala, Punjab 147002, India.
| |
Collapse
|
52
|
Wang M, Guo Z, Wang S. Regulation of cystathionine γ-lyase in mammalian cells by hypoxia. Biochem Genet 2013; 52:29-37. [PMID: 23852134 DOI: 10.1007/s10528-013-9624-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Accepted: 04/16/2013] [Indexed: 12/25/2022]
Abstract
Hydrogen sulfide (H2S), an endogenous signaling molecule in mammalian cells, shows a variety of biological effects. Cystathionine γ-lyase (CSE) is a key enzyme in the trans-sulfuration pathway responsible for the production of endogenous H2S. Whether CSE expression is regulated by hypoxia in mammalian cells remains largely unknown. This study revealed that these regulatory effects changed with time at transcriptional and post-transcriptional levels. Hypoxia regulated CSE expression in mammalian cells in a complex manner; CSE transcription went through a down-regulation and recovery period, while CSE mRNA and protein levels increased during hypoxia. Taken together, the results suggest that CSE can respond to hypoxia through transcriptional and post-transcriptional regulation, and CSE expression can be up-regulated by hypoxia to a certain extent. Therefore, the up-regulation of CSE expression during hypoxia may be useful for increasing the production and concentration of H2S in mammalian cells and indirectly protecting cells from hypoxia.
Collapse
Affiliation(s)
- Maoxian Wang
- College of Life Sciences and Technology, Tongji University, 1239 Siping Road, Shanghai, 200092, China
| | | | | |
Collapse
|
53
|
Huang C, Kan J, Liu X, Ma F, Tran BH, Zou Y, Wang S, Zhu YZ. Cardioprotective effects of a novel hydrogen sulfide agent-controlled release formulation of S-propargyl-cysteine on heart failure rats and molecular mechanisms. PLoS One 2013; 8:e69205. [PMID: 23874913 PMCID: PMC3706411 DOI: 10.1371/journal.pone.0069205] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Accepted: 06/05/2013] [Indexed: 01/29/2023] Open
Abstract
Objective Heart failure (HF) is one of the most serious diseases worldwide. S-propargyl-cysteine (SPRC), a novel modulator of endogenous hydrogen sulfide, is proved to be able to protect against acute myocardial ischemia. In order to produce more stable and sustainable hydrogen sulfide, we used controlled release formulation of SPRC (CR-SPRC) to elucidate possible cardioprotective effects on HF rats and investigate involved mechanisms on apoptosis and oxidation. Methods Left coronary artery was occluded to induce HF model of rat. The survival rats were randomly divided into 7 groups after 24 hours and treated with drugs for 6 weeks. Echocardiographic indexes were recorded to determine cardiac function. TTC staining was performed to determine infarct size. Plasmatic level of hydrogen sulfide was detected by modified sulfide electrode. Activity of enzyme and expression of protein were determined by colorimetry and Western blot, respectively. Results The cardioprotective effects of CR-SPRC on HF rats were confirmed by significant reduction of infarct size and improvement of cardiac function, with better effects compared to normal SPRC. CR-SPRC modulated antioxidant defenses by preserving levels of GSH, CAT and SOD and reducing CK leakage. In addition, CR-SPRC elevated ratio of Bcl-2/Bax and inhibited activity of caspases to protect against myocardial apoptosis. The cardioprotective effects of CR-SPRC were mediated by hydrogen sulfide. Conclusions All experiment data indicated cardioprotective effects of CR-SPRC on HF rats. More importantly, CR-SPRC exerted better effects than normal SPRC in all respects, providing a new perspective on hydrogen sulfide-mediated drug therapy.
Collapse
Affiliation(s)
- Chengrong Huang
- School of Pharmacy, Fudan University, Shanghai, Shanghai, China
| | - Juntao Kan
- School of Pharmacy & Institutes of Biomedical Sciences, Fudan University, Shanghai, Shanghai, China
| | - Xu Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
| | - Fenfen Ma
- School of Pharmacy, Fudan University, Shanghai, Shanghai, China
| | - Ba Hieu Tran
- School of Pharmacy, Fudan University, Shanghai, Shanghai, China
| | - Yunzeng Zou
- Zhongshan Hospital & Institutes of Biomedical Sciences, Fudan University, Shanghai, Shanghai, China
| | - Shujun Wang
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, Liaoning, China
- * E-mail: (YZZ); (SW)
| | - Yi Zhun Zhu
- School of Pharmacy & Institutes of Biomedical Sciences, Fudan University, Shanghai, Shanghai, China
- * E-mail: (YZZ); (SW)
| |
Collapse
|
54
|
Bos EM, Wang R, Snijder PM, Boersema M, Damman J, Fu M, Moser J, Hillebrands JL, Ploeg RJ, Yang G, Leuvenink HGD, van Goor H. Cystathionine γ-lyase protects against renal ischemia/reperfusion by modulating oxidative stress. J Am Soc Nephrol 2013; 24:759-70. [PMID: 23449534 DOI: 10.1681/asn.2012030268] [Citation(s) in RCA: 144] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous gasotransmitter with physiologic functions similar to nitric oxide and carbon monoxide. Exogenous treatment with H2S can induce a reversible hypometabolic state, which can protect organs from ischemia/reperfusion injury, but whether cystathionine γ-lyase (CSE), which produces endogenous H2S, has similar protective effects is unknown. Here, human renal tissue revealed abundant expression of CSE, localized to glomeruli and the tubulointerstitium. Compared with wild-type mice, CSE knockout mice had markedly reduced renal production of H2S, and CSE deficiency associated with increased damage and mortality after renal ischemia/reperfusion injury. Treatment with exogenous H2S rescued CSE knockout mice from the injury and mortality associated with renal ischemia. In addition, overexpression of CSE in vitro reduced the amount of reactive oxygen species produced during stress. Last, the level of renal CSE mRNA at the time of organ procurement positively associated with GFR 14 days after transplantation. In summary, these results suggest that CSE protects against renal ischemia/reperfusion injury, likely by modulating oxidative stress through the production of H2S.
Collapse
Affiliation(s)
- Eelke M Bos
- Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
55
|
Liu C, Guo W, Maerz S, Gu X, Zhu Y. 3,5-Dimethoxy-4-(3-(2-carbonyl-ethyldisulfanyl)-propionyl)-benzoic acid 4-guanidino-butyl ester: a novel twin drug that prevents primary cardiac myocytes from hypoxia-induced apoptosis. Eur J Pharmacol 2013; 700:118-26. [PMID: 23305837 DOI: 10.1016/j.ejphar.2012.11.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 11/06/2012] [Accepted: 11/13/2012] [Indexed: 01/24/2023]
Abstract
Leonurine possesses cardioprotective effects in myocardial ischemia due to its anti-apoptotic properties. However, the process to isolate and purify leonurine is difficult, because of its low content in the Herb Leonuri and its impurity. Moreover, the high dosage used indicates low potency of leonurine. To overcome these defects, we had synthesized a novel twin drug of leonurine, 3,5-dimethoxy-4-(3-(2-carbonyl-ethyldisulfanyl)-propionyl)-benzoic acid 4-guanidino-butyl ester (compound 2). In this paper, we focused on investigating the cardioprotective effect and underlying mechanisms of compound 2. Our data showed that cell viability was significantly increased in a dose-dependent manner and the levels of lactate dehydrogenase (LDH) and creatine kinase (CK) were also significantly attenuated in the compound 2-treated group. In addition, we observed the cardioprotective effects by Hoechst 33258 nucleus staining, JC-1 staining, Annexin V-FITC/PI staining and transmission electron microscopy. Compound 2 inhibited apoptosis by reducing the ratio of Bcl-2/Bax, decreasing cleaved-caspase-3 expression and enhancing the phosphorylation of Akt. Furthermore, the phosphorylation effect of compound 2 was reversed by LY294002 the phosphatidylinositol-3-kinase (PI3K) inhibitor from happening. We concluded that compound 2 played a cardioprotective role in hypoxia-induced primary cardiac myocytes apoptosis partly via modulating the PI3K/Akt pathway at a 10-fold lower concentration than leonurine.
Collapse
Affiliation(s)
- Chunhua Liu
- Department of Pharmacology, School of Pharmacy and Institute of Biomedical Sciences, Fudan University, Shanghai 201203, China
| | | | | | | | | |
Collapse
|
56
|
Bhatia M. Role of hydrogen sulfide in the pathology of inflammation. SCIENTIFICA 2012; 2012:159680. [PMID: 24278674 PMCID: PMC3820548 DOI: 10.6064/2012/159680] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 10/03/2012] [Indexed: 06/02/2023]
Abstract
Hydrogen sulfide (H2S) is a well-known toxic gas that is synthesized in the human body from the amino acids cystathionine, homocysteine, and cysteine by the action of 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 biological mediator. Imbalances in H2S have also been shown to be associated with various disease conditions. However, defining the precise pathophysiology of H2S is proving to be a complex challenge. Recent research in our laboratory has shown H2S as a novel mediator of inflammation and work in several groups worldwide is currently focused on determining the role of H2S in inflammation. H2S has been implicated in different inflammatory conditions, such as acute pancreatitis, sepsis, joint inflammation, and chronic obstructive pulmonary disease (COPD). Active research on the role of H2S in inflammation will unravel the pathophysiology of its actions in inflammatory conditions and may help develop novel therapeutic approaches for several, as yet incurable, disease conditions.
Collapse
Affiliation(s)
- Madhav Bhatia
- Department of Pathology, University of Otago, P.O. Box 4345, Christchurch 8140, New Zealand
| |
Collapse
|
57
|
Gu X, Zhu YZ. Therapeutic applications of organosulfur compounds as novel hydrogen sulfide donors and/or mediators. Expert Rev Clin Pharmacol 2012; 4:123-33. [PMID: 22115353 DOI: 10.1586/ecp.10.129] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Hydrogen sulfide, once considered as toxic gas, is now recognized as an important biological mediator. The deficiency of hydrogen sulfide could lead to various pathological changes, such as arterial and pulmonary hypertension, Alzheimer's disease, gastric mucosal injury and liver cirrhosis. However, excessive production of hydrogen sulfide, by using inorganic hydrogen sulfide donors such as NaHS, may contribute to the pathogenesis of inflammatory diseases, septic shock, cerebral stroke and mental retardation in patients with Down syndrome. Therefore, an increasing interest in organic molecules that are capable of regulating the formation of hydrogen sulfide has extended in recent years. Allium vegetables are one natural source of organic sulfur-containing compounds and have been widely investigated regarding their therapeutic applications, and it has been proven that the ingredients of garlic, such as diallyl disulfide, diallyl trisulfide and S-ally cysteine act as hydrogen sulfide donors or mediators in pharmaceutical studies. In addition, S-propargyl cysteine (ZYZ-802) and S-propyl cysteine, two synthetic cysteine analogs, have been examined and could be used to treat ischemic heart disease via modulation of the hydrogen sulfide pathway. In addition, drugs containing hydrogen sulfide-releasing moieties have been synthesized and widely reported in recent years, such as S-nonsteroidal anti-inflammatory drugs and the derivative of Lawesson's reagents, which exhibit varied biological effects in experiments. As cystathionine β-synthase and cystathionine γ-lyase are the enzymes that are able to catalyze the production of endogenous hydrogen sulfide from cysteine, their inhibitors, such as dl-propylargylglycine and β-cyanoalanine, have been frequently used in studies on the biological mechanism of hydrogen sulfide. All these hydrogen sulfide donors, mediators and inhibitors have provided useful tools in the research of a variety of biological effects and are promising drug candidates of hydrogen sulfide.
Collapse
Affiliation(s)
- Xianfeng Gu
- Fudan University, Shanghai 200032, People's Republic of China
| | | |
Collapse
|
58
|
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
|
59
|
Pan LL, Liu XH, Gong QH, Yang HB, Zhu YZ. Role of cystathionine γ-lyase/hydrogen sulfide pathway in cardiovascular disease: a novel therapeutic strategy? Antioxid Redox Signal 2012; 17:106-18. [PMID: 22017202 PMCID: PMC3342562 DOI: 10.1089/ars.2011.4349] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2011] [Accepted: 10/21/2011] [Indexed: 12/31/2022]
Abstract
SIGNIFICANCE Hydrogen sulfide (H(2)S) has traditionally been considered a toxic environmental pollutant. In the late 1990s, the presumed solely harmful role of H(2)S has been challenged because H(2)S may also be involved in the maintenance and preservation of cardiovascular homeostasis. RECENT ADVANCES The production of endogenous H(2)S has been attributed to three key enzymes, cystathionine γ-lyase (CSE), cystathionine β-synthase, and 3-mercaptopyruvate sulfurtransferase. The recognition of H(2)S as the third gaseous signaling molecule has stimulated research on a multitude of pathophysiologic events in the cardiovascular system. In particular, important roles in cardiovascular disorder processes are ascribed to the CSE/H(2)S pathway, such as atherosclerosis, myocardial infarction, hypertension, and shock. CRITICAL ISSUES Many biological activities and molecular mechanisms of H(2)S in the cardiovascular system have been demonstrated in studies using different tools, such as the genetic overexpression of CSE, the direct administration of H(2)S donors, or the use of H(2)S-releasing pro-drugs. Unfortunately, the role of the CSE/H(2)S pathway in cardiovascular disease remains controversial in numerous areas, and many questions regarding the gaseous molecule still remain unanswered. FUTURE DIRECTIONS Advances in basic research indicate that the CSE/H(2)S pathway may provide potential therapeutic targets for treating cardiovascular disorders. But the molecular targets of H(2)S still need to be identified.
Collapse
Affiliation(s)
- Li Long Pan
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Xin Hua Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Qi Hai Gong
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - He Bei Yang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yi Zhun Zhu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, China
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
| |
Collapse
|
60
|
Zheng YT, Zhu JH, Ma G, Zhu Q, Yang P, Tan B, Zhang JL, Shen HX, Xu JL, Zhu YZ, Cai WM. Preclinical assessment of the distribution, metabolism, and excretion of S-propargyl-cysteine, a novel H2S donor, in Sprague-Dawley rats. Acta Pharmacol Sin 2012; 33:839-44. [PMID: 22543704 DOI: 10.1038/aps.2012.15] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
AIM To study the distribution, metabolism and excretion of S-propargyl-cysteine (SPRC), a novel hydrogen sulfide (H2S) donor, after oral administration in rats. METHODS Adult Sprague-Dawley rats were used. The tissue distribution of [(35)S] SPRC-derived radioactivity was measured using a liquid scintillation counter. The plasma protein binding of SPRC was examined using 96-well equilibrium dialysis. The excretion of SPRC in urine, bile and feces was analyzed using the LC-MS/MS method. The major metabolites in rat biomatrices were identified using MRM information-dependent, acquisition-enhanced product ion (MRM-IDA-EPI) scans on API 4000QTrap system. RESULTS After oral administration of [(35)S]-SPRC at a dose of 75 mg/kg, [(35)S] SPRC-derived radioactivity displayed broad biological distribution in various tissues of rats, including its target organs (heart and brain) with the highest in kidney. On the other hand, the binding of SPRC to human, rat and dog plasma protein was low. Only 2.18% ± 0.61% and 0.77% ± 0.61% of the total SPRC administered was excreted unchanged in the bile and urine. However, neither intact SPRC nor its metabolites were detected in rat feces. The major metabolic pathway in vivo (rat bile, urine, and plasma) was N-acetylation. CONCLUSION The preliminary results suggest that SPRC possesses acceptable pharmacokinetic properties in rats.
Collapse
|
61
|
Zhu HM, Deng L. Evaluation of cardiomyocyte hypoxia injury models for the pharmacological study in vitro. PHARMACEUTICAL BIOLOGY 2012; 50:167-74. [PMID: 22085279 DOI: 10.3109/13880209.2011.583255] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
CONTEXT Traditional Herbal Medicine (THM) has many advantages that make it a promising choice for the treatment of ischemic heart disease (IHD). To study the mechanism of IHDs or pharmacological actions of THM, many hypoxia-induced cardiomyocyte injury models have been established. Radix Salvia miltorrhiza (Danshen) was used as a representative of THM. Danshen is a famous medicinal herb widely applied in Asia to relieve ischemic cardiovascular diseases. OBJECTIVE To investigate the effects of various hypoxic conditions and discuss a suitable hypoxia model, cell viability, apoptosis, release of myocardial injury markers, and mRNA levels of target genes were tested for the first time. MATERIALS AND METHODS Radix Salvia miltorrhiza (Danshen) was purchased from a GMP-compliant producer and both its preparation method and quality control were standardized. Cellular status, such as cell viability, apoptosis, releases of myocardial injury markers, and the mRNA level of target gene were tested by 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) method, biochemical analyzer, flow cytometry, Hoechst 33258 staining, and real-time PCR, respectively. RESULTS Based on our data, we found a treppe response of cardiomyocyte in the hypoxic condition and suggested that 8 h in 2% O2 might be a suitable condition for in vitro pharmacological study of cardiomyocytes. DISCUSSION AND CONCLUSIONS Our findings outlined more extended and in-depth capability of cardiomyocyte suffering from hypoxia, and might be of particular interest due to the high prevalence of THM pharmacological study.
Collapse
Affiliation(s)
- Hong-Ming Zhu
- Division of Stem Cell and Tissue Engineering, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | | |
Collapse
|
62
|
Novel anti-diabetic effect of SCM-198 via inhibiting the hepatic NF-κB pathway in db/db mice. Biosci Rep 2012; 32:185-95. [PMID: 21859425 DOI: 10.1042/bsr20110017] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
There are reports of early evidence that suggest the involvement of chronic low-grade inflammation in the pathogenesis of Type 2 diabetes. Thus, substances that have effects in reducing inflammation could be potential drugs for Type 2 diabetes. Leonurine (4-guanidino-n-butyl syringate; SCM-198) is an alkaloid in HL (Herba leonuri), which was reported to possess anti-inflammatory properties. We hypothesize that SCM-198 may have beneficial effects on Type 2 diabetes. In the present study, we attempted to test this hypothesis by evaluating the anti-diabetic effect of SCM-198 and the possible underlying mechanisms of its effects in db/db mice. SCM-198 (50, 100 and 200 mg/kg of body weight), pioglitazone (50 mg/kg of body weight, as a positive control) or 1% CMC-Na (sodium carboxymethylcellulose) were administered to the db/db or db/m mice once daily for 3 weeks. After 3 weeks, SCM-198 (200 mg/kg of body weight) treatment significantly reduced the fasting blood glucose level and increased the plasma insulin concentration in the db/db mice, meanwhile it significantly lowered the plasma TAG (triacylglycerol) concentration and increased the HDL (high-density lipoprotein)-cholesterol concentration. Moreover, the dysregulated transcription of the hepatic glucose metabolic enzymes, including GK (glucokinase), G6Pase (glucose-6-phosphatase) and PEPCK (phosphoenolpyruvate carboxykinase), was recovered by an Akt-dependent pathway. The pro-inflammatory mediators {such as TNFα (tumour necrosis factor α), IL (interleukin)-6, IL-1β, degradation of IκB [inhibitor of NF-κB (nuclear factor-κB)] α and thereafter activation of NF-κB} were reversed by SCM-198 treatment in the db/db mice. The present study provides first evidence that SCM-198 exhibits anti-inflammatory activity and has an ameliorating effect on diabetic symptoms via inhibiting of NF-κB/IKK (IκB kinase) pathway. Consequently, we suggest that SCM-198 may be a prospective agent for prevention and/or moderation of the progress of Type 2 diabetes.
Collapse
|
63
|
Sidhapuriwala JN, Hegde A, Ang AD, Zhu YZ, Bhatia M. Effects of S-propargyl-cysteine (SPRC) in caerulein-induced acute pancreatitis in mice. PLoS One 2012; 7:e32574. [PMID: 22396778 PMCID: PMC3291555 DOI: 10.1371/journal.pone.0032574] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2011] [Accepted: 01/27/2012] [Indexed: 01/23/2023] Open
Abstract
Hydrogen sulfide (H2S), a novel gaseous messenger, is synthesized endogenously from L-cysteine by two pyridoxal-5′-phosphate-dependent enzymes, cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE). S-propargyl-cysteine (SPRC) is a slow H2S releasing drug that provides cysteine, a substrate of CSE. The present study was aimed to investigate the effects of SPRC in an in vivo model of acute pancreatitis (AP) in mice. AP was induced in mice by hourly caerulein injections (50 µg/kg) for 10 hours. Mice were treated with SPRC (10 mg/kg) or vehicle (distilled water). SPRC was administered either 12 h before or 3 h before the induction of pancreatitis. Mice were sacrificed 1 h after the last caerulein injection. Blood, pancreas and lung tissues were collected and processed to measure the plasma amylase, plasma H2S, myeloperoxidase (MPO) activities and cytokine levels in pancreas and lung. The results revealed that significant reduction of inflammation, both in pancreas and lung was associated with SPRC given 3 h prior to the induction of AP. Furthermore, the beneficial effects of SPRC were associated with reduction of pancreatic and pulmonary pro-inflammatory cytokines and increase of anti-inflammatory cytokine. SPRC administered 12 h before AP induction did not cause significant improvement in pancreatic and lung inflammation. Plasma H2S concentration showed significant difference in H2S levels between control, vehicle and SPRC (administered 3 h before AP) treatment groups. In conclusion, these data provide evidence for protective effects of SPRC in AP possibly by virtue of its slow release of endogenous H2S.
Collapse
Affiliation(s)
| | - Akhil Hegde
- Department of Pathology, University of Otago, Christchurch, New Zealand
| | - Abel D. Ang
- Department of Pathology, University of Otago, Christchurch, New Zealand
| | - Yi Zhun Zhu
- Department of Pharmacology, National University of Singapore, Singapore, Singapore
- Department of Pharmacology, Fudan University, Shanghai, China
| | - Madhav Bhatia
- Department of Pathology, University of Otago, Christchurch, New Zealand
- * E-mail:
| |
Collapse
|
64
|
Pan LL, Liu XH, Zheng HM, Yang HB, Gong QH, Zhu YZ. S-propargyl-cysteine, a novel hydrogen sulfide-modulated agent, attenuated tumor necrosis factor-α-induced inflammatory signaling and dysfunction in endothelial cells. Int J Cardiol 2012; 155:327-32. [PMID: 22240755 DOI: 10.1016/j.ijcard.2011.12.059] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2011] [Revised: 12/11/2011] [Accepted: 12/17/2011] [Indexed: 01/14/2023]
|
65
|
Zheng Y, Xu J, Ma G, Zhang J, Zhu Q, Liu H, Zhang P, Zhu Y, Cai W. Bioavailability and pharmacokinetics of S-propargyl-L-cysteine, a novel cardioprotective agent, after single and multiple doses in Beagle dogs. Xenobiotica 2011; 42:304-9. [PMID: 21961645 DOI: 10.3109/00498254.2011.617848] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
As a novel hydrogen sulfide-modulated agent, S-propargyl-L-cysteine (SPRC) is proven to be a potent cardioprotective candidate. Bioavailability and pharmacokinetics of SPRC (20 mg/kg) in beagle dogs after oral and intravenous administrations were investigated in this study. Plasma concentrations of SPRC were measured by a LC-MS/MS method. Intravenous administration of SPRC (single dose) to beagle dogs gave a mean plasma half-life of 14.7 h, mean clearance of 0.4 ml min⁻¹ kg⁻¹ and mean apparent volume of distribution of 0.56 L/kg. Single oral administration was completely, fast absorbed (T(max)= 0.33 ± 0.20 h) with a mean absolute availability of 112% and mean plasma half-life of 16.5 h. Multiple oral administration (once daily for 10 consecutive days) of SPRC to dogs resulted in steady state plasma drug concentration being reached after seven doses and didn't cause obvious accumulation. No significant difference was found between the single and multiple pharmacokinetic parameters.
Collapse
Affiliation(s)
- Yuanting Zheng
- Department of Clinical Pharmacy, School of Pharmacy, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | |
Collapse
|
66
|
Leonurine-cysteine analog conjugates as a new class of multifunctional anti-myocardial ischemia agent. Eur J Med Chem 2011; 46:3996-4009. [DOI: 10.1016/j.ejmech.2011.05.073] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2011] [Revised: 05/30/2011] [Accepted: 05/31/2011] [Indexed: 11/24/2022]
|
67
|
Development of a boron-dipyrromethene-Cu2+ ensemble based colorimetric probe toward hydrogen sulfide in aqueous media. Tetrahedron Lett 2011. [DOI: 10.1016/j.tetlet.2011.07.004] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
|
68
|
MA K, Liu Y, Zhu Q, Liu CH, Duan JL, Tan BKH, Zhu YZ. H2S donor, S-propargyl-cysteine, increases CSE in SGC-7901 and cancer-induced mice: evidence for a novel anti-cancer effect of endogenous H2S? PLoS One 2011; 6:e20525. [PMID: 21738579 PMCID: PMC3124470 DOI: 10.1371/journal.pone.0020525] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2011] [Accepted: 05/02/2011] [Indexed: 01/03/2023] Open
Abstract
Background S-propargyl-cysteine (SPRC), an H2S donor, is a structural analogue of S-allycysteine (SAC). It was investigated for its potential anti-cancer effect on SGC-7901 gastric cancer cells and the possible mechanisms that may be involved. Methods and Findings SPRC treatment significantly decreased cell viability, suppressed the proliferation and migration of SPRC-7901 gastric cancer cells, was pro-apoptotic as well as caused cell cycle arrest at the G1/S phase. In an in vivo study, intra-peritoneal injection of 50 mg/kg and 100 mg/kg of SPRC significantly reduced tumor weights and tumor volumes of gastric cancer implants in nude mice, with a tumor growth inhibition rate of 40–75%. SPRC also induced a pro-apoptotic effect in cancer tissues and elevated the expressions of p53 and Bax in tumors and cells. SPRC treatment also increased protein expression of cystathione-γ-lyase (CSE) in cells and tumors, and elevated H2S levels in cell culture media, plasma and tumoral CSE activity of gastric cancer-induced nude mice by 2, 2.3 and 1.4 fold, respectively. Most of the anti-cancer functions of SPRC on cells and tumors were significantly suppressed by PAG, an inhibitor of CSE activity. Conclusions Taken together, the results of our study provide insights into a novel anti-cancer effect of H2S as well as of SPRC on gastric cancer through inducing the activity of a new target, CSE.
Collapse
Affiliation(s)
- Kaium MA
- Department of Pharmacology, School of Pharmacy and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Yan Liu
- Department of Cardiovascular Medicine, Ruijin Hospital of Shanghai Jiaotong University, Shanghai, China
| | - Qing Zhu
- Department of Pharmacology, School of Pharmacy and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chun-hua Liu
- Department of Pharmacology, School of Pharmacy and Institute of Biomedical Sciences, Fudan University, Shanghai, China
| | - Jun-Li Duan
- Department of Gerontology, Xinhua Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Benny K-H. Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yi Zhun Zhu
- Department of Pharmacology, School of Pharmacy and Institute of Biomedical Sciences, Fudan University, Shanghai, China
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- * E-mail:
| |
Collapse
|
69
|
Li K, Wang H, Cheng L, Wang M, Zhu R, Wang SL. Characterization of transient species produced from laser flash photolysis of a new cardioprotective drug: S-propargyl-cysteine. J Photochem Photobiol A Chem 2011. [DOI: 10.1016/j.jphotochem.2011.02.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
70
|
Zheng Y, Liu H, Ma G, Yang P, Zhang L, Gu Y, Zhu Q, Shao T, Zhang P, Zhu Y, Cai W. Determination of S-propargyl-cysteine in rat plasma by mixed-mode reversed-phase and cation-exchange HPLC–MS/MS method and its application to pharmacokinetic studies. J Pharm Biomed Anal 2011; 54:1187-91. [DOI: 10.1016/j.jpba.2010.11.027] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2010] [Revised: 11/11/2010] [Accepted: 11/22/2010] [Indexed: 11/29/2022]
|
71
|
S-Propargyl-cysteine (SPRC) attenuated lipopolysaccharide-induced inflammatory response in H9c2 cells involved in a hydrogen sulfide-dependent mechanism. Amino Acids 2011; 41:205-15. [PMID: 21308383 DOI: 10.1007/s00726-011-0834-1] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2010] [Accepted: 01/11/2011] [Indexed: 01/28/2023]
Abstract
The present study attempts to investigate the effects of S-propargyl-cysteine (SPRC), a sulfur-containing amino acid, on lipopolysaccharide (LPS)-induced inflammatory response in H9c2 cardiac myocytes. We found that SPRC prevented nuclear factor-κB (NF-κB) activation assessed by NF-κB p65 phosphorylation and IκBα degradation, suppressed LPS-induced extracellular signal-regulated kinase 1/2 (ERK1/2) phosphorylation and intracellular reactive oxygen species (ROS) production. Furthermore, incubation of H9c2 cells with SPRC induced phosphorylation of Akt in a time- and concentration-dependent manner. In addition, SPRC attenuated LPS-induced mRNA and protein expression of tumor necrosis factor-α (TNF-α), and mRNA expression of intercellular adhesion molecule-1 (ICAM-1) and inducible nitric oxide synthase (iNOS). The effects of SPRC were abolished by cystathionine γ-lyase [CSE-an enzyme that synthesizes hydrogen sulfide (H(2)S)] inhibitor, DL: -propargylglycine (PAG), SPRC-induced Akt phosphorylation and TNF-α release was also abolished by the phosphoinositide 3-kinase (PI3K) inhibitor LY294002. Furthermore, SPRC also increased LPS-induced down-regulation expression of CSE and H(2)S level in H9c2 cells. PAG abolished SPRC-induced up-regulation of H(2)S level. Therefore, we concluded that SPRC produced an anti-inflammatory effect in LPS-stimulated H9c2 cells partly through the CSE/H(2)S pathway by impairing IκBα/NF-κB signaling and by activating PI3K/Akt signaling pathway.
Collapse
|
72
|
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: 8.1] [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
|
73
|
Liu C, Gu X, Zhu YZ. Synthesis and biological evaluation of novel leonurine–SPRC conjugate as cardioprotective agents. Bioorg Med Chem Lett 2010; 20:6942-6. [DOI: 10.1016/j.bmcl.2010.09.135] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 09/26/2010] [Accepted: 09/28/2010] [Indexed: 11/26/2022]
|
74
|
Szabó G, Veres G, Radovits T, Gero D, Módis K, Miesel-Gröschel C, Horkay F, Karck M, Szabó C. Cardioprotective effects of hydrogen sulfide. Nitric Oxide 2010; 25:201-10. [PMID: 21094267 DOI: 10.1016/j.niox.2010.11.001] [Citation(s) in RCA: 98] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2010] [Revised: 11/04/2010] [Accepted: 11/08/2010] [Indexed: 01/19/2023]
Abstract
The gaseous mediator hydrogen sulfide (H(2)S) is synthesized mainly by cystathionine γ-lyase in the heart and plays a role in the regulation of cardiovascular homeostasis. Here we first overview the state of the art in the literature on the cardioprotective effects of H(2)S in various models of cardiac injury. Subsequently, we present original data showing the beneficial effects of parenteral administration of a donor of H(2)S on myocardial and endothelial function during reperfusion in a canine experimental model of cardiopulmonary bypass. Overview of the literature demonstrates that various formulations of H(2)S exert cardioprotective effects in cultured cells, isolated hearts and various rodent and large animal models of regional or global myocardial ischemia and heart failure. In addition, the production of H(2)S plays a role in myocardial pre- and post-conditioning responses. The pathways implicated in the cardioprotective action of H(2)S are multiple and involve K(ATP) channels, regulation of mitochondrial respiration, and regulation of cytoprotective genes such as Nrf-2. In the experimental part of the current article, we demonstrate the cardioprotective effects of H(2)S in a canine model of cardiopulmonary bypass surgery. Anesthetized dogs were subjected hypothermic cardiopulmonary bypass with 60 min of hypothermic cardiac arrest in the presence of either saline (control, n=8), or H(2)S infusion (1 mg/kg/h for 2 h). Left ventricular hemodynamic variables (via combined pressure-volume-conductance catheter) as well as coronary blood flow, endothelium-dependent vasodilatation to acetylcholine and endothelium-independent vasodilatation to sodium nitroprusside were measured at baseline and after 60 min of reperfusion. Ex vivo vascular function and high-energy phosphate contents were also measured. H(2)S led to a significantly better recovery of preload recruitable stroke work (p<0.05) after 60 min of reperfusion. Coronary blood flow was also significantly higher in the H(2)S group (p<0.05). While the vasodilatory response to sodium nitroprusside was similar in both groups, acetylcholine resulted in a significantly higher increase in coronary blood flow in the H(2)S-treated group (p<0.05) both in vivo and ex vivo. Furthermore, high-energy phosphate contents were better preserved in the H(2)S group. Additionally, the cytoprotective effects of H(2)S were confirmed also using in vitro cell culture experiments in H9c2 cardiac myocytes exposed to hypoxia and reoxygenation or to the cytotoxic oxidant hydrogen peroxide. Thus, therapeutic administration of H(2)S exerts cardioprotective effects in a variety of experimental models, including a significant improvement of the recovery of myocardial and endothelial function in a canine model of cardiopulmonary bypass with hypothermic cardiac arrest.
Collapse
Affiliation(s)
- Gábor Szabó
- Department of Cardiac Surgery, University of Heidelberg, Im Neuenheiemer Feld 110, 69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Protective effects of novel single compound, Hirsutine on hypoxic neonatal rat cardiomyocytes. Eur J Pharmacol 2010; 650:290-7. [PMID: 20940014 DOI: 10.1016/j.ejphar.2010.09.057] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Revised: 09/16/2010] [Accepted: 09/20/2010] [Indexed: 11/20/2022]
Abstract
Uncaria rhynchophylla is a traditional Chinese herb that has been applied in China for treatment of ailments of the cardiovascular system, but little is known about its active constituents and effect in cardiomyocytes. In present study, we investigated the cardioprotective effect of 0.1μΜ, 1μΜ and 10μΜ Hirsutine isolated from the methanolic extracts of Uncaria rhynchophylla by high performance liquid chromatography (HPLC) on neonatal rat cardiomyocytes treated with hypoxia to determine the mechanism underlying the protective effect with regard to cardiac anti-oxidant enzymes and apoptosis genes. Hirsutine significantly increased the viability of cardiomyocytes injured by hypoxia. Gene expression levels of proapoptotic genes (Bax, Fas and caspase-3) were significantly downregulated compared with the hypoxic control group (P<0.05), whereas the expression level of Bcl-2 was upregulated following Hirsutine treatment (P<0.05). Correspondingly, Hirsutine treatment increased Bcl-2 protein level and decreased Bax protein level. Assay investigating cardiac anti-oxidant enzymes provided further evidence for the protective effect of Hirsutine, as indicated by the induction of the anti-oxidant enzymes superoxide dismutase. The results of present study suggest that the mechanism of action of Hirsutine in hypoxic neonatal rat cardiomyocytes may be related to its anti-oxidant and anti-apoptotic properties. This may open an avenue for developing novel candidate compounds with cardioprotectiveeffect from unique Chinese plant.
Collapse
|
76
|
S-propargyl-cysteine (ZYZ-802), a sulphur-containing amino acid, attenuates beta-amyloid-induced cognitive deficits and pro-inflammatory response: involvement of ERK1/2 and NF-κB pathway in rats. Amino Acids 2010; 40:601-10. [DOI: 10.1007/s00726-010-0685-1] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2010] [Accepted: 06/30/2010] [Indexed: 01/01/2023]
|
77
|
Predmore BL, Lefer DJ. Development of hydrogen sulfide-based therapeutics for cardiovascular disease. J Cardiovasc Transl Res 2010; 3:487-98. [PMID: 20628909 DOI: 10.1007/s12265-010-9201-y] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2010] [Accepted: 06/21/2010] [Indexed: 10/19/2022]
Abstract
The physiological role of the gaseous signaling molecule hydrogen sulfide (H(2)S) was first realized in the mid-1990s with the work of Abe and Kimura. Since then, it has become evident that this endogenous gas is extremely important in the homeostasis of the cardiovascular system and the pathogenesis of cardiovascular disease. Several biotechnology companies have developed and are developing H(2)S-based therapeutic compounds, and there are ongoing clinical trials investigating the therapeutic potential of H(2)S. Several organic and chemical compounds that are known H(2)S donors have the potential to be developed into effective H(2)S-based therapeutic agents. This review will provide a historical and current perspective on the role(s) of H(2)S in the cardiovascular system and the current state of development and future outlook of H(2)S-based therapies for cardiovascular disease.
Collapse
Affiliation(s)
- Benjamin L Predmore
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, 550 Peachtree Street, NE, Atlanta, GA 30308, USA
| | | |
Collapse
|
78
|
Gong QH, Wang Q, Pan LL, Liu XH, Huang H, Zhu YZ. Hydrogen sulfide attenuates lipopolysaccharide-induced cognitive impairment: A pro-inflammatory pathway in rats. Pharmacol Biochem Behav 2010; 96:52-8. [DOI: 10.1016/j.pbb.2010.04.006] [Citation(s) in RCA: 66] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2010] [Revised: 03/31/2010] [Accepted: 04/08/2010] [Indexed: 10/19/2022]
|
79
|
Calvert JW, Coetzee WA, Lefer DJ. Novel insights into hydrogen sulfide--mediated cytoprotection. Antioxid Redox Signal 2010; 12:1203-17. [PMID: 19769484 PMCID: PMC2864658 DOI: 10.1089/ars.2009.2882] [Citation(s) in RCA: 243] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hydrogen sulfide (H(2)S) is a colorless, water soluble, flammable gas that has the characteristic smell of rotten eggs. Like other members of the gasotransmitter family (nitric oxide and carbon monoxide), H(2)S has traditionally been considered to be a highly toxic gas and environmental hazard. However, much like for nitric oxide and carbon monoxide, the initial negative perception of H(2)S has evolved with the discovery that H(2)S is produced enzymatically in mammals under normal conditions. As a result of this discovery, there has been a great deal of work to elucidate the physiological role of H(2)S. H(2)S is now recognized to be cytoprotective in various models of cellular injury. Specifically, it has been demonstrated that the acute administration of H(2)S, either prior to ischemia or at reperfusion, significantly ameliorates in vitro or in vivo myocardial and hepatic ischemia-reperfusion injury. These studies have also demonstrated a cardioprotective role for endogenous H(2)S. This review article summarizes the current body of evidence demonstrating the cytoprotective effects of H(2)S with an emphasis on the cardioprotective effects. This review also provides a detailed description of the current signaling mechanisms shown to be responsible for these cardioprotective actions.
Collapse
Affiliation(s)
- John W Calvert
- Department of Surgery, Division of Cardiothoracic Surgery, Emory University School of Medicine, Atlanta, Georgia, USA
| | | | | |
Collapse
|
80
|
Wang Q, Wang XL, Liu HR, Rose P, Zhu YZ. Protective effects of cysteine analogues on acute myocardial ischemia: novel modulators of endogenous H(2)S production. Antioxid Redox Signal 2010; 12:1155-65. [PMID: 19842912 DOI: 10.1089/ars.2009.2947] [Citation(s) in RCA: 88] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The current study was designed to evaluate the pharmacologic effects of three novel cysteine-containing compounds: S-propyl-l-cysteine (SPC), S-allyl-l-cysteine (SAC), and S-propargyl-l-cysteine (SPRC) on H(2)S production and antioxidant defenses in an acute myocardial infarction (MI) rat model. The enzymatic activities of superoxide dismutase (SOD), catalase (CAT), and glutathione peroxidase (GPx), as well as glutathione redox status and malonaldehyde (MDA) content, also were determined. All three compounds were found to preserve SOD and GPx activities and also tissue GSH levels while reducing the formation of the lipid peroxidation product MDA in ventricular tissues. With immunfluorescence assays, we observed the expression of CSE and Mn-SOD. The morphologic changes of the cardiac cells are seen with both light and electron microscopy. The corresponding pathologic alterations were characterized mainly as loss of adherence between cardiac myocytes and swollen or ruptured mitochondria at the ultrastructural level. Propargylglycine, a selective inhibitor of CSE, abolished the protective effects of each compound used in the current model. Our study provides novel evidence that SPC, SAC, and SPRC have cardioprotective effects in MI by reducing the deleterious effects of oxidative stress by modulating the endogenous levels of H(2)S and preserving the activities of antioxidant defensive enzymes like SOD.
Collapse
Affiliation(s)
- Qian Wang
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | | | | | | | | |
Collapse
|
81
|
Abstract
Vertebrate cardiorespiratory homeostasis is inextricably dependent upon specialized cells that provide feedback on oxygen status in the tissues, blood, and on occasion, environment. These "oxygen sensing" cells include chemoreceptors and oxygen-sensitive chromaffin cells that initiate cardiorespiratory reflexes, vascular smooth muscle cells that adjust perfusion to metabolism or ventilation, and other cells that condition themselves in response to episodic hypoxia. Identification of how these cells sense oxygen and transduce this into the appropriate physiological response has enormous clinical applicability, but despite intense research there is no consensus regarding the initial hypoxia-effector coupling mechanism. This review examines an alternative mechanism of oxygen sensing using oxidation of endogenously produced hydrogen sulfide (H(2)S) as the O(2)-sensitive couple. Support for this hypothesis includes the similarity of effects of hypoxia and H(2)S on a variety of tissues, augmentation of hypoxic responses by precursors of H(2)S production and their inhibition by inhibitors of H(2)S synthesis, and the rapid consumption of H(2)S by O(2) in the range of intracellular/mitochondrial Po(2). These studies also indicate that, under normoxic conditions, it is doubtful that free H(2)S has longer than a transient existence in tissue or extracellular fluid.
Collapse
Affiliation(s)
- Kenneth R Olson
- Indiana University School of Medicine, South Bend Center, South Bend, Indiana 46617, USA.
| | | |
Collapse
|