1
|
Zampas P, Li Z, Katsouda A, Varela A, Psarras S, Davos CH, Lefer DJ, Papapetropoulos A. Protective role of 3-mercaptopyruvate sulfurtransferase (MPST) in the development of metabolic syndrome and vascular inflammation. Pharmacol Res 2025; 211:107542. [PMID: 39667544 DOI: 10.1016/j.phrs.2024.107542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 11/19/2024] [Accepted: 12/06/2024] [Indexed: 12/14/2024]
Abstract
Metabolic syndrome (MetS) is a cluster of metabolic abnormalities that occur concurrently and increase the risk of cardiovascular disease. 3-mercaptopyruvate sulfurtransferase (MPST) is a cysteine-catabolizing enzyme that yields pyruvate and hydrogen sulfide (H2S) and plays a central role in the regulation of energy homeostasis. Herein, we seek to investigate the role of MPST/H2S in MetS and its cardiovascular consequences using a mouse model of the disease. Mice were fed a high-fat diet (HFD) for 15 weeks to induce obesity and hyperglycemia and administrated a nitric oxide synthase inhibitor, during the last 5 weeks to induce hypertension and MetS. This model caused a mild left ventricular (LV) diastolic dysfunction and vascular endothelial dysfunction. Free H2S and sulfane-sulfur levels were decreased in the aorta, but unaltered in the heart. Also, downregulation of MPST and thiosulfate sulfuretransferase (TST) were observed in the aorta. Global deletion of Mpst (Mpst-/-) resulted in increased body weight and greater glucose intolerance in mice with MetS, without affecting their blood pressure, and caused an upregulation of genes involved in immune responses in the vasculature suggestive of T-cell infiltration and activation. Pharmacological restoration of H2S levels ameliorated the comorbidities of MetS; GYY4137 administration reduced body weight and blood pressure, attenuated cardiac fibrosis and improved glucose handling and endothelium-dependent relaxation. In conclusion, this study found that reduced MPST/H2S exacerbates the pathological changes associated with MetS and contributes to vascular inflammation. H2S supplementation emerges as a potential therapeutic approach to treat the abnormalities associated with MetS.
Collapse
Affiliation(s)
- Paraskevas Zampas
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, Greece; Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Zhen Li
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Antonia Katsouda
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, Greece; British Heart Foundation Centre of Research Excellence, Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, England, UK
| | - Aimilia Varela
- Cardiovascular Laboratory, Biomedical Research Foundation Academy of Athens, Greece
| | - Stelios Psarras
- Center of Basic Research, Biomedical Research Foundation Academy of Athens, Greece
| | - Constantinos H Davos
- Cardiovascular Laboratory, Biomedical Research Foundation Academy of Athens, Greece
| | - David J Lefer
- Department of Cardiac Surgery, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Andreas Papapetropoulos
- Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation Academy of Athens, Greece; Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece.
| |
Collapse
|
2
|
Zhang G, Sun X, Zhang D, Zhang X, Yu K. SerpinA3 Promotes Myocardial Infarction in Rat and Cell-based Models. Mol Biotechnol 2025; 67:92-103. [PMID: 38006519 DOI: 10.1007/s12033-023-00982-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 11/06/2023] [Indexed: 11/27/2023]
Abstract
This study aimed to examine the role and molecular mechanism of the nuclear factor κB (NFκB)/serine protease inhibitor A3 (SerpinA3) interaction in myocardial ischemia-reperfusion (IR) injury. First, a rat model for myocardial ischemia-reperfusion injury was established, using 2,3,5-triphenyltetrazolium chloride to measure the size of the myocardial infarction. Pathological variations in myocardial tissue were detected using hematoxylin-eosin staining. Flow cytometry and terminal deoxynucleotidyl transferase biotin-dUTP nick end labeling (TUNEL) staining were used to measure cell death in the rat model. The SerpinA3 mRNA and protein expressions in the myocardium of IR-model rats were remarkably higher than those in the control group. Furthermore, the oxidative, inflammatory, and apoptotic activities of the myocardial tissue of SerpinA3-knockdown (KD) rats were significantly improved compared to those in the WT group. SerpinA3-KD also contributed to the recovery of cardiac function in IR-model rats. Additionally, silencing of SerpinA3 inhibited p65 phosphorylation in myocardial tissues and reduced H2O2-induced inflammation, oxidative stress, and apoptosis in myocardial cells. The expression of SerpinA3 increased in myocardial tissue after IR stimulation. Knockdown of SerpinA3 can deactivate NF-κB and reduce inflammation, oxidative stress, and apoptosis in vivo and in vitro, thereby lessening myocardial injury caused by IR. In conclusion, SerpinA3 promotes myocardial infarction in rat and cell-based models by activating NF-κB. However, the mechanism by which increased Serpina3 expression causes downstream NF-κB activation to mediate the proposed, pathological effects in myocardial IR injury remain untested and worthy of future investigations.
Collapse
Affiliation(s)
- Gang Zhang
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1, Huanghe Xi Lu, Huaiyin District, Huaian City, 223300, Jiangsu Province, China
| | - Xiaofeng Sun
- Department of Pediatric, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, 223300, Jiangsu, China
| | - Dongying Zhang
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1, Huanghe Xi Lu, Huaiyin District, Huaian City, 223300, Jiangsu Province, China
| | - Xiwen Zhang
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1, Huanghe Xi Lu, Huaiyin District, Huaian City, 223300, Jiangsu Province, China
| | - Kun Yu
- Department of Cardiology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1, Huanghe Xi Lu, Huaiyin District, Huaian City, 223300, Jiangsu Province, China.
| |
Collapse
|
3
|
Li A, Wu S, Li Q, Wang Q, Chen Y. Elucidating the Molecular Pathways and Therapeutic Interventions of Gaseous Mediators in the Context of Fibrosis. Antioxidants (Basel) 2024; 13:515. [PMID: 38790620 PMCID: PMC11117599 DOI: 10.3390/antiox13050515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 04/13/2024] [Accepted: 04/22/2024] [Indexed: 05/26/2024] Open
Abstract
Fibrosis, a pathological alteration of the repair response, involves continuous organ damage, scar formation, and eventual functional failure in various chronic inflammatory disorders. Unfortunately, clinical practice offers limited treatment strategies, leading to high mortality rates in chronic diseases. As part of investigations into gaseous mediators, or gasotransmitters, including nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), numerous studies have confirmed their beneficial roles in attenuating fibrosis. Their therapeutic mechanisms, which involve inhibiting oxidative stress, inflammation, apoptosis, and proliferation, have been increasingly elucidated. Additionally, novel gasotransmitters like hydrogen (H2) and sulfur dioxide (SO2) have emerged as promising options for fibrosis treatment. In this review, we primarily demonstrate and summarize the protective and therapeutic effects of gaseous mediators in the process of fibrosis, with a focus on elucidating the underlying molecular mechanisms involved in combating fibrosis.
Collapse
Affiliation(s)
- Aohan Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian 116622, China; (A.L.); (S.W.); (Q.L.)
| | - Siyuan Wu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian 116622, China; (A.L.); (S.W.); (Q.L.)
| | - Qian Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian 116622, China; (A.L.); (S.W.); (Q.L.)
| | - Qianqian Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian 116622, China; (A.L.); (S.W.); (Q.L.)
- Engineering Technology Research Center for The Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian 116622, China
| | - Yingqing Chen
- Chronic Disease Research Center, Medical College, Dalian University, Dalian 116622, China; (A.L.); (S.W.); (Q.L.)
- Engineering Technology Research Center for The Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian 116622, China
| |
Collapse
|
4
|
Łoboda A, Dulak J. Cardioprotective Effects of Hydrogen Sulfide and Its Potential Therapeutic Implications in the Amelioration of Duchenne Muscular Dystrophy Cardiomyopathy. Cells 2024; 13:158. [PMID: 38247849 PMCID: PMC10814317 DOI: 10.3390/cells13020158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Hydrogen sulfide (H2S) belongs to the family of gasotransmitters and can modulate a myriad of biological signaling pathways. Among others, its cardioprotective effects, through antioxidant, anti-inflammatory, anti-fibrotic, and proangiogenic activities, are well-documented in experimental studies. Cardiorespiratory failure, predominantly cardiomyopathy, is a life-threatening complication that is the number one cause of death in patients with Duchenne muscular dystrophy (DMD). Although recent data suggest the role of H2S in ameliorating muscle wasting in murine and Caenorhabditis elegans models of DMD, possible cardioprotective effects have not yet been addressed. In this review, we summarize the current understanding of the role of H2S in animal models of cardiac dysfunctions and cardiac cells. We highlight that DMD may be amenable to H2S supplementation, and we suggest H2S as a possible factor regulating DMD-associated cardiomyopathy.
Collapse
Affiliation(s)
- Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7 Street, 30-387 Kraków, Poland;
| | | |
Collapse
|
5
|
Hu J, Xu Z, Liao D, Jiang Y, Pu H, Wu Z, Xu X, Zhao Z, Liu J, Lu X, Liu X, Li B. An H 2 S-BMP6 Dual-Loading System with Regulating Yap/Taz and Jun Pathway for Synergistic Critical Limb Ischemia Salvaging Therapy. Adv Healthc Mater 2023; 12:e2301316. [PMID: 37531238 DOI: 10.1002/adhm.202301316] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Revised: 07/12/2023] [Indexed: 08/04/2023]
Abstract
Critical limb ischemia, the final course of peripheral artery disease, is characterized by an insufficient supply of blood flow and excessive oxidative stress. H2 S molecular therapy possesses huge potential for accelerating revascularization and scavenging intracellular reactive oxygen species (ROS). Moreover, it is found that BMP6 is the most significantly up-expressed secreted protein-related gene in HUVECs treated with GYY4137, a H2 S donor, based on the transcriptome analysis. Herein, a UIO-66-NH2 @GYY4137@BMP6 co-delivery nanoplatform to strengthen the therapeutic effects of limb ischemia is developed. The established UIO-66-NH2 @GYY4137@BMP6 nanoplatform exerts its proangiogenic and anti-oxidation functions by regulating key pathways. The underlying molecular mechanisms of UIO-66-NH2 @GYY4137@BMP6 dual-loading system lie in the upregulation of phosphorylated YAP/TAZ and Jun to promote HUVECs proliferation and downregulation of phosphorylated p53/p21 to scavenge excessive ROS. Meanwhile, laser-doppler perfusion imaging (LDPI), injury severity evaluation, and histological analysis confirm the excellent therapeutic effects of UIO-66-NH2 @GYY4137@BMP6 in vivo. This work may shed light on the treatment of critical limb ischemia by regulating YAP, Jun, and p53 signaling pathways based on gas-protein synergistic therapy.
Collapse
Affiliation(s)
- Jiateng Hu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhijue Xu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Donghui Liao
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, 523700, China
| | - Yihong Jiang
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Hongji Pu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhaoyu Wu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Xintong Xu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Zhen Zhao
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Jianqiang Liu
- The First Dongguan Affiliated Hospital, Guangdong Medical University, Zhanjiang, 523700, China
| | - Xinwu Lu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Xiaobing Liu
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Bo Li
- Department of Vascular Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
- Vascular Centre of Shanghai Jiao Tong University, Shanghai, 200011, China
| |
Collapse
|
6
|
Zhang Y, Jing M, Cai C, Zhu S, Zhang C, Wang Q, Zhai Y, Ji X, Wu D. Role of hydrogen sulphide in physiological and pathological angiogenesis. Cell Prolif 2022; 56:e13374. [PMID: 36478328 PMCID: PMC9977675 DOI: 10.1111/cpr.13374] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2022] [Revised: 11/08/2022] [Accepted: 11/21/2022] [Indexed: 12/12/2022] Open
Abstract
The role of hydrogen sulphide (H2 S) in angiogenesis has been widely demonstrated. Vascular endothelial growth factor (VEGF) plays an important role in H2 S-induced angiogenesis. H2 S promotes angiogenesis by upregulating VEGF via pro-angiogenic signal transduction. The involved signalling pathways include the mitogen-activated protein kinase pathway, phosphoinositide-3 kinase pathway, nitric oxide (NO) synthase/NO pathway, signal transducer and activator of transcription 3 (STAT3) pathway, and adenosine triphosphate (ATP)-sensitive potassium (KATP ) channels. H2 S has been shown to contribute to tumour angiogenesis, diabetic wound healing, angiogenesis in cardiac and cerebral ischaemic tissues, and physiological angiogenesis during the menstrual cycle and pregnancy. Furthermore, H2 S can exert an anti-angiogenic effect by inactivating Wnt/β-catenin signalling or blocking the STAT3 pathway in tumours. Therefore, H2 S plays a double-edged sword role in the process of angiogenesis. The regulation of H2 S production is a promising therapeutic approach for angiogenesis-associated diseases. Novel H2 S donors and/or inhibitors can be developed in the treatment of angiogenesis-dependent diseases.
Collapse
Affiliation(s)
- Yan‐Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Mi‐Rong Jing
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Chun‐Bo Cai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Shuai‐Gang Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Chao‐Jing Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Qi‐Meng Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina
| | - Yuan‐Kun Zhai
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,School of StomatologyHenan UniversityKaifengHenanChina
| | - Xin‐Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina,Kaifeng Key Laboratory of Infection and Biological Safety, School of Basic Medical SciencesHenan UniversityKaifengHenanChina
| | - Dong‐Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical SciencesHenan UniversityKaifengHenanChina,Kaifeng Municipal Key Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular MedicineHenan UniversityKaifengHenanChina,School of StomatologyHenan UniversityKaifengHenanChina
| |
Collapse
|
7
|
Miljkovic JL, Burger N, Gawel JM, Mulvey JF, Norman AAI, Nishimura T, Tsujihata Y, Logan A, Sauchanka O, Caldwell ST, Morris JL, Prime TA, Warrington S, Prudent J, Bates GR, Aksentijević D, Prag HA, James AM, Krieg T, Hartley RC, Murphy MP. Rapid and selective generation of H 2S within mitochondria protects against cardiac ischemia-reperfusion injury. Redox Biol 2022; 55:102429. [PMID: 35961099 PMCID: PMC9382561 DOI: 10.1016/j.redox.2022.102429] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 07/22/2022] [Accepted: 08/01/2022] [Indexed: 02/02/2023] Open
Abstract
Mitochondria-targeted H2S donors are thought to protect against acute ischemia-reperfusion (IR) injury by releasing H2S that decreases oxidative damage. However, the rate of H2S release by current donors is too slow to be effective upon administration following reperfusion. To overcome this limitation here we develop a mitochondria-targeted agent, MitoPerSulf that very rapidly releases H2S within mitochondria. MitoPerSulf is quickly taken up by mitochondria, where it reacts with endogenous thiols to generate a persulfide intermediate that releases H2S. MitoPerSulf is acutely protective against cardiac IR injury in mice, due to the acute generation of H2S that inhibits respiration at cytochrome c oxidase thereby preventing mitochondrial superoxide production by lowering the membrane potential. Mitochondria-targeted agents that rapidly generate H2S are a new class of therapy for the acute treatment of IR injury.
Collapse
Affiliation(s)
- Jan Lj Miljkovic
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK
| | - Nils Burger
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK
| | - Justyna M Gawel
- School of Chemistry, University of Glasgow, Glasgow, G12 8QQ, UK
| | - John F Mulvey
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | | | - Takanori Nishimura
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK; Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 251-8555, Japan
| | - Yoshiyuki Tsujihata
- Innovative Biology Laboratories, Neuroscience Drug Discovery Unit, Takeda Pharmaceutical Company Limited, 251-8555, Japan
| | - Angela Logan
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK
| | - Olga Sauchanka
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | | | - Jordan L Morris
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK
| | - Tracy A Prime
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK
| | | | - Julien Prudent
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK
| | - Georgina R Bates
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK
| | - Dunja Aksentijević
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, United Kingdom
| | - Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK; Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Andrew M James
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | | | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, CB2 0XY, UK; Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
8
|
Yang YW, Deng NH, Tian KJ, Liu LS, Wang Z, Wei DH, Liu HT, Jiang ZS. Development of hydrogen sulfide donors for anti-atherosclerosis therapeutics research: Challenges and future priorities. Front Cardiovasc Med 2022; 9:909178. [PMID: 36035922 PMCID: PMC9412017 DOI: 10.3389/fcvm.2022.909178] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/28/2022] [Indexed: 11/16/2022] Open
Abstract
Hydrogen sulfide (H2S), a gas transmitter found in eukaryotic organisms, plays an essential role in several physiological processes. H2S is one of the three primary biological gas transmission signaling mediators, along with nitric oxide and carbon monoxide. Several animal and in vitro experiments have indicated that H2S can prevent coronary endothelial mesenchymal transition, reduce the expression of endothelial cell adhesion molecules, and stabilize intravascular plaques, suggesting its potential role in the treatment of atherosclerosis (AS). H2S donors are compounds that can release H2S under certain circumstances. Development of highly targeted H2S donors is a key imperative as these can allow for in-depth evaluation of the anti-atherosclerotic effects of exogenous H2S. More importantly, identification of an optimal H2S donor is critical for the creation of H2S anti-atherosclerotic prodrugs. In this review, we discuss a wide range of H2S donors with anti-AS potential along with their respective transport pathways and design-related limitations. We also discuss the utilization of nano-synthetic technologies to manufacture H2S donors. This innovative and effective design example sheds new light on the production of highly targeted H2S donors.
Collapse
Affiliation(s)
- Ye-Wei Yang
- Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, Institute of Pathogenic Biology, Hengyang Medical School, University of South China, Hengyang, China
| | - Nian-Hua Deng
- Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Kai-Jiang Tian
- Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Lu-Shan Liu
- Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Zuo Wang
- Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Dang-Heng Wei
- Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Hui-Ting Liu
- Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, Institute of Cardiovascular Disease, University of South China, Hengyang, China
| | - Zhi-Sheng Jiang
- Key Laboratory for Arteriosclerosis of Hunan Province, Hengyang Medical College, Institute of Cardiovascular Disease, University of South China, Hengyang, China
- *Correspondence: Zhi-Sheng Jiang
| |
Collapse
|
9
|
Zhu C, Liu Q, Li X, Wei R, Ge T, Zheng X, Li B, Liu K, Cui R. Hydrogen sulfide: A new therapeutic target in vascular diseases. Front Endocrinol (Lausanne) 2022; 13:934231. [PMID: 36034427 PMCID: PMC9399516 DOI: 10.3389/fendo.2022.934231] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Hydrogen sulfide (H2S) is one of most important gas transmitters. H2S modulates many physiological and pathological processes such as inflammation, oxidative stress and cell apoptosis that play a critical role in vascular function. Recently, solid evidence show that H2S is closely associated to various vascular diseases. However, specific function of H2S remains unclear. Therefore, in this review we systemically summarized the role of H2S in vascular diseases, including hypertension, atherosclerosis, inflammation and angiogenesis. In addition, this review also outlined a novel therapeutic perspective comprising crosstalk between H2S and smooth muscle cell function. Therefore, this review may provide new insight inH2S application clinically.
Collapse
Affiliation(s)
- Cuilin Zhu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Qing Liu
- Department of Cardiovascular Medicine, University of Tokyo, Tokyo, Japan
| | - Xin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Ran Wei
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Xiufen Zheng
- Department of Surgery, Western University, London, ON, Canada
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
| | - Kexiang Liu
- Department of Cardiovascular Surgery, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Ranji Cui, ; Kexiang Liu,
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Ranji Cui, ; Kexiang Liu,
| |
Collapse
|
10
|
Wu D, Gu Y, Zhu D. Cardioprotective effects of hydrogen sulfide in attenuating myocardial ischemia‑reperfusion injury (Review). Mol Med Rep 2021; 24:875. [PMID: 34726247 DOI: 10.3892/mmr.2021.12515] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 08/05/2021] [Indexed: 11/05/2022] Open
Abstract
Ischemic heart disease is one of the major causes of cardiovascular‑related mortality worldwide. Myocardial ischemia can be attenuated by reperfusion that restores the blood supply. However, injuries occur during blood flow restoration that induce cardiac dysfunction, which is known as myocardial ischemia‑reperfusion injury (MIRI). Hydrogen sulfide (H2S), the third discovered endogenous gasotransmitter in mammals (after NO and CO), participates in various pathophysiological processes. Previous in vitro and in vivo research have revealed the protective role of H2S in the cardiovascular system that render it useful in the protection of the myocardium against MIRI. The cardioprotective effects of H2S in attenuating MIRI are summarized in the present review.
Collapse
Affiliation(s)
- Dan Wu
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Yijing Gu
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, P.R. China
| | - Deqiu Zhu
- Tongji Hospital, School of Medicine, Tongji University, Shanghai 200065, P.R. China
| |
Collapse
|
11
|
Chirindoth SS, Cancarevic I. Role of Hydrogen Sulfide in the Treatment of Fibrosis. Cureus 2021; 13:e18088. [PMID: 34692303 PMCID: PMC8525665 DOI: 10.7759/cureus.18088] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 09/18/2021] [Indexed: 12/29/2022] Open
Abstract
Hydrogen sulfide (H2S) is a biological gas, the abnormal metabolism of which has associations with the pathogenesis of fibrosis. The purpose of this paper was to determine the potential of H2S in the prevention and treatment of fibrosis. The data is obtained mainly from articles found in the PubMed database using the keywords “fibrosis” and “hydrogen sulfide,” limiting the results to those published within the last 10 years. Some additional resources have also been used, such as books and articles within journals. Evidence of decreased H2S enzyme levels in animal models with fibrotic diseases has been found. The protective role of H2S has been validated by the administration of exogenous H2S donors in animal models with fibrosis. It is also evident that H2S is involved in complex signaling pathways and ion channels that inhibit fibrosis development. These findings support the role of H2S in the treatment of a variety of fibrotic diseases. A randomized controlled trial in fibrosis patients comparing the efficacy of exogenous H2S and placebo in addition to standard of care can be implemented to validate this further.
Collapse
Affiliation(s)
- Swathy S Chirindoth
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| | - Ivan Cancarevic
- Internal Medicine, California Institute of Behavioral Neurosciences & Psychology, Fairfield, USA
| |
Collapse
|
12
|
Mendiola PJ, Naik JS, Bosc LVG, Gardiner AS, Birg A, Kanagy NL. Hydrogen Sulfide Actions in the Vasculature. Compr Physiol 2021; 11:2467-2488. [PMID: 34558672 PMCID: PMC11758848 DOI: 10.1002/cphy.c200036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Hydrogen sulfide (H2 S) is a small, gaseous molecule with poor solubility in water that is generated by multiple pathways in many species including humans. It acts as a signaling molecule in many tissues with both beneficial and pathological effects. This article discusses its many actions in the vascular system and the growing evidence of its role to regulate vascular tone, angiogenesis, endothelial barrier function, redox, and inflammation. Alterations in some disease states are also discussed including potential roles in promoting tumor growth and contributions to the development of metabolic disease. © 2021 American Physiological Society. Compr Physiol 11:1-22, 2021.
Collapse
Affiliation(s)
| | - Jay S. Naik
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | | | - Amy S. Gardiner
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Aleksandr Birg
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| | - Nancy L. Kanagy
- University of New Mexico Health Sciences Center, Albuquerque, New Mexico, USA
| |
Collapse
|
13
|
Chen Y, Yuan S, Cao Y, Kong G, Jiang F, Li Y, Wang Q, Tang M, Zhang Q, Wang Q, Liu L. Gasotransmitters: Potential Therapeutic Molecules of Fibrotic Diseases. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:3206982. [PMID: 34594474 PMCID: PMC8478550 DOI: 10.1155/2021/3206982] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 08/31/2021] [Indexed: 02/06/2023]
Abstract
Fibrosis is defined as the pathological progress of excessive extracellular matrix (ECM), such as collagen, fibronectin, and elastin deposition, as the regenerative capacity of cells cannot satisfy the dynamic repair of chronic damage. The well-known features of tissue fibrosis are characterized as the presence of excessive activated and proliferated fibroblasts and the differentiation of fibroblasts into myofibroblasts, and epithelial cells undergo the epithelial-mesenchymal transition (EMT) to expand the number of fibroblasts and myofibroblasts thereby driving fibrogenesis. In terms of mechanism, during the process of fibrosis, the activations of the TGF-β signaling pathway, oxidative stress, cellular senescence, and inflammatory response play crucial roles in the activation and proliferation of fibroblasts to generate ECM. The deaths due to severe fibrosis account for almost half of the total deaths from various diseases, and few treatment strategies are available for the prevention of fibrosis as yet. Recently, numerous studies demonstrated that three well-defined bioactive gasotransmitters, including nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S), generally exhibited anti-inflammatory, antioxidative, antiapoptotic, and antiproliferative properties. Besides these effects, a number of studies have reported that low-dose exogenous and endogenous gasotransmitters can delay and interfere with the occurrence and development of fibrotic diseases, including myocardial fibrosis, idiopathic pulmonary fibrosis, liver fibrosis, renal fibrosis, diabetic diaphragm fibrosis, and peritoneal fibrosis. Furthermore, in animal and clinical experiments, the inhalation of low-dose exogenous gas and intraperitoneal injection of gaseous donors, such as SNAP, CINOD, CORM, SAC, and NaHS, showed a significant therapeutic effect on the inhibition of fibrosis through modulating the TGF-β signaling pathway, attenuating oxidative stress and inflammatory response, and delaying the cellular senescence, while promoting the process of autophagy. In this review, we first demonstrate and summarize the therapeutic effects of gasotransmitters on diverse fibrotic diseases and highlight their molecular mechanisms in the process and development of fibrosis.
Collapse
Affiliation(s)
- Yingqing Chen
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Shuo Yuan
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002 Jilin Province, China
| | - Yuying Cao
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Guangyao Kong
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Feng Jiang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - You Li
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Qi Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Minli Tang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Qinggao Zhang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
- Key Laboratory of Natural Medicines of the Changbai Mountain, Ministry of Education, College of Pharmacy, Yanbian University, Yanji, 133002 Jilin Province, China
| | - Qianqian Wang
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| | - Liping Liu
- Chronic Disease Research Center, Medical College, Dalian University, Dalian, 116622 Liaoning, China
- Engineering Technology Research Center for the Utilization of Functional Components of Organic Natural Products, Dalian University, Dalian, 116622 Liaoning, China
| |
Collapse
|
14
|
Testai L, Brancaleone V, Flori L, Montanaro R, Calderone V. Modulation of EndMT by Hydrogen Sulfide in the Prevention of Cardiovascular Fibrosis. Antioxidants (Basel) 2021; 10:antiox10060910. [PMID: 34205197 PMCID: PMC8229400 DOI: 10.3390/antiox10060910] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/23/2021] [Accepted: 06/01/2021] [Indexed: 02/06/2023] Open
Abstract
Endothelial mesenchymal transition (EndMT) has been described as a fundamental process during embryogenesis; however, it can occur also in adult age, underlying pathological events, including fibrosis. Indeed, during EndMT, the endothelial cells lose their specific markers, such as vascular endothelial cadherin (VE-cadherin), and acquire a mesenchymal phenotype, expressing specific products, such as α-smooth muscle actin (α-SMA) and type I collagen; moreover, the integrity of the endothelium is disrupted, and cells show a migratory, invasive and proliferative phenotype. Several stimuli can trigger this transition, but transforming growth factor (TGF-β1) is considered the most relevant. EndMT can proceed in a canonical smad-dependent or non-canonical smad-independent manner and ultimately regulate gene expression of pro-fibrotic machinery. These events lead to endothelial dysfunction and atherosclerosis at the vascular level as well as myocardial hypertrophy and fibrosis. Indeed, EndMT is the mechanism which promotes the progression of cardiovascular disorders following hypertension, diabetes, heart failure and also ageing. In this scenario, hydrogen sulfide (H2S) has been widely described for its preventive properties, but its role in EndMT is poorly investigated. This review is focused on the evaluation of the putative role of H2S in the EndMT process.
Collapse
Affiliation(s)
- Lara Testai
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (L.F.); (V.C.)
- Interdepartmental Center of Ageing, University of Pisa, 56126 Pisa, Italy
- Correspondence:
| | - Vincenzo Brancaleone
- Department of Science, University of Basilicata, 85100 Potenza, Italy; (V.B.); (R.M.)
| | - Lorenzo Flori
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (L.F.); (V.C.)
| | - Rosangela Montanaro
- Department of Science, University of Basilicata, 85100 Potenza, Italy; (V.B.); (R.M.)
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, 56126 Pisa, Italy; (L.F.); (V.C.)
- Interdepartmental Center of Ageing, University of Pisa, 56126 Pisa, Italy
| |
Collapse
|
15
|
LaPenna KB, Polhemus DJ, Doiron JE, Hidalgo HA, Li Z, Lefer DJ. Hydrogen Sulfide as a Potential Therapy for Heart Failure-Past, Present, and Future. Antioxidants (Basel) 2021; 10:485. [PMID: 33808673 PMCID: PMC8003444 DOI: 10.3390/antiox10030485] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/05/2021] [Accepted: 03/17/2021] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous, gaseous signaling molecule that plays a critical role in cardiac and vascular biology. H2S regulates vascular tone and oxidant defenses and exerts cytoprotective effects in the heart and circulation. Recent studies indicate that H2S modulates various components of metabolic syndrome, including obesity and glucose metabolism. This review will discuss studies exhibiting H2S -derived cardioprotective signaling in heart failure with reduced ejection fraction (HFrEF). We will also discuss the role of H2S in metabolic syndrome and heart failure with preserved ejection fraction (HFpEF).
Collapse
Affiliation(s)
- Kyle B. LaPenna
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - David J. Polhemus
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Jake E. Doiron
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Hunter A. Hidalgo
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - Zhen Li
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | - David J. Lefer
- Cardiovascular Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA; (K.B.L.); (D.J.P.); (J.E.D.); (H.A.H.); (Z.L.)
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| |
Collapse
|
16
|
Ciccone V, Genah S, Morbidelli L. Endothelium as a Source and Target of H 2S to Improve Its Trophism and Function. Antioxidants (Basel) 2021; 10:antiox10030486. [PMID: 33808872 PMCID: PMC8003673 DOI: 10.3390/antiox10030486] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 12/18/2022] Open
Abstract
The vascular endothelium consists of a single layer of squamous endothelial cells (ECs) lining the inner surface of blood vessels. Nowadays, it is no longer considered as a simple barrier between the blood and vessel wall, but a central hub to control blood flow homeostasis and fulfill tissue metabolic demands by furnishing oxygen and nutrients. The endothelium regulates the proper functioning of vessels and microcirculation, in terms of tone control, blood fluidity, and fine tuning of inflammatory and redox reactions within the vessel wall and in surrounding tissues. This multiplicity of effects is due to the ability of ECs to produce, process, and release key modulators. Among these, gasotransmitters such as nitric oxide (NO) and hydrogen sulfide (H2S) are very active molecules constitutively produced by endotheliocytes for the maintenance and control of vascular physiological functions, while their impairment is responsible for endothelial dysfunction and cardiovascular disorders such as hypertension, atherosclerosis, and impaired wound healing and vascularization due to diabetes, infections, and ischemia. Upregulation of H2S producing enzymes and administration of H2S donors can be considered as innovative therapeutic approaches to improve EC biology and function, to revert endothelial dysfunction or to prevent cardiovascular disease progression. This review will focus on the beneficial autocrine/paracrine properties of H2S on ECs and the state of the art on H2S potentiating drugs and tools.
Collapse
|
17
|
Wang WL, Ge TY, Chen X, Mao Y, Zhu YZ. Advances in the Protective Mechanism of NO, H 2S, and H 2 in Myocardial Ischemic Injury. Front Cardiovasc Med 2020; 7:588206. [PMID: 33195476 PMCID: PMC7661694 DOI: 10.3389/fcvm.2020.588206] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/28/2020] [Indexed: 12/30/2022] Open
Abstract
Myocardial ischemic injury is among the top 10 leading causes of death from cardiovascular diseases worldwide. Myocardial ischemia is caused mainly by coronary artery occlusion or obstruction. It usually occurs when the heart is insufficiently perfused, oxygen supply to the myocardium is reduced, and energy metabolism in the myocardium is abnormal. Pathologically, myocardial ischemic injury generates a large number of inflammatory cells, thus inducing a state of oxidative stress. This sharp reduction in the number of normal cells as a result of apoptosis leads to organ and tissue damage, which can be life-threatening. Therefore, effective methods for the treatment of myocardial ischemic injury and clarification of the underlying mechanisms are urgently required. Gaseous signaling molecules, such as NO, H2S, H2, and combined gas donors, have gradually become a focus of research. Gaseous signaling molecules have shown anti-apoptotic, anti-oxidative and anti-inflammatory effects as potential therapeutic agents for myocardial ischemic injury in a large number of studies. In this review, we summarize and discuss the mechanism underlying the protective effect of gaseous signaling molecules on myocardial ischemic injury.
Collapse
Affiliation(s)
| | | | - Xu Chen
- Guilin Medical College, Guilin, China
| | - Yicheng Mao
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| | - Yi-Zhun Zhu
- Guilin Medical College, Guilin, China.,Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China.,State Key Laboratory of Quality Research in Chinese Medicine and School of Pharmacy, Macau University of Science and Technology, Macau, China
| |
Collapse
|
18
|
Denoix N, McCook O, Ecker S, Wang R, Waller C, Radermacher P, Merz T. The Interaction of the Endogenous Hydrogen Sulfide and Oxytocin Systems in Fluid Regulation and the Cardiovascular System. Antioxidants (Basel) 2020; 9:E748. [PMID: 32823845 PMCID: PMC7465147 DOI: 10.3390/antiox9080748] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/10/2020] [Accepted: 08/11/2020] [Indexed: 12/11/2022] Open
Abstract
The purpose of this review is to explore the parallel roles and interaction of hydrogen sulfide (H2S) and oxytocin (OT) in cardiovascular regulation and fluid homeostasis. Their interaction has been recently reported to be relevant during physical and psychological trauma. However, literature reports on H2S in physical trauma and OT in psychological trauma are abundant, whereas available information regarding H2S in psychological trauma and OT in physical trauma is much more limited. This review summarizes recent direct and indirect evidence of the interaction of the two systems and their convergence in downstream nitric oxide-dependent signaling pathways during various types of trauma, in an effort to better understand biological correlates of psychosomatic interdependencies.
Collapse
Affiliation(s)
- Nicole Denoix
- Clinic for Psychosomatic Medicine and Psychotherapy, Ulm University Medical Center, 89081 Ulm, Germany;
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Oscar McCook
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Sarah Ecker
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Rui Wang
- Faculty of Science, York University, Toronto, ON M3J 1P3, Canada;
| | - Christiane Waller
- Department of Psychosomatic Medicine and Psychotherapy, Nuremberg General Hospital, Paracelsus Medical University, 90419 Nuremberg, Germany;
| | - Peter Radermacher
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| | - Tamara Merz
- Institute for Anesthesiological Pathophysiology and Process Engineering, Ulm University Medical Center, 89081 Ulm, Germany; (S.E.); (P.R.); (T.M.)
| |
Collapse
|
19
|
Ellmers LJ, Templeton EM, Pilbrow AP, Frampton C, Ishii I, Moore PK, Bhatia M, Richards AM, Cameron VA. Hydrogen Sulfide Treatment Improves Post-Infarct Remodeling and Long-Term Cardiac Function in CSE Knockout and Wild-Type Mice. Int J Mol Sci 2020; 21:4284. [PMID: 32560137 PMCID: PMC7352717 DOI: 10.3390/ijms21124284] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 06/10/2020] [Accepted: 06/12/2020] [Indexed: 12/14/2022] Open
Abstract
Hydrogen sulfide (H2S) is recognized as an endogenous gaseous signaling molecule generated by cystathionine γ-lyase (CSE) in cardiovascular tissues. H2S up-regulation has been shown to reduce ischemic injury, and H2S donors are cardioprotective in rodent models when administered concurrent with myocardial ischemia. We evaluated the potential utility of H2S therapy in ameliorating cardiac remodeling with administration delayed until 2 h post-infarction in mice with or without cystathionine γ-lyase gene deletion (CSE-/-). The slow-release H2S donor, GYY4137, was administered from 2 h after surgery and daily for 28 days following myocardial infarction (MI) induced by coronary artery ligation, comparing responses in CSE-/- with wild-type (WT) mice (n = 5-10/group/genotype). Measures of cardiac function and expression of key genes associated with cardiac hypertrophy, fibrosis, and apoptosis were documented in atria, ventricle, and kidney tissues. Post-MI GYY4137 administration reduced infarct area and restored cardiac function, accompanied by reduction of the elevated ventricular expression of genes mediating cardiac remodeling to near-normal levels. Few differences between WT and CSE-/- mice were observed, except CSE-/- mice had higher blood pressures, and higher atrial Mir21a expression across all treatment groups. These findings suggest endogenous CSE gene deletion does not substantially exacerbate the long-term response to MI. Moreover, the H2S donor GYY4137 administered after onset of MI preserves cardiac function and protects against adverse cardiac remodeling in both WT and CSE-deficient mice.
Collapse
Affiliation(s)
- Leigh J. Ellmers
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8140, New Zealand; (L.J.E.); (E.M.T.); (A.P.P.); (C.F.); (A.M.R.)
| | - Evelyn M. Templeton
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8140, New Zealand; (L.J.E.); (E.M.T.); (A.P.P.); (C.F.); (A.M.R.)
| | - Anna P. Pilbrow
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8140, New Zealand; (L.J.E.); (E.M.T.); (A.P.P.); (C.F.); (A.M.R.)
| | - Chris Frampton
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8140, New Zealand; (L.J.E.); (E.M.T.); (A.P.P.); (C.F.); (A.M.R.)
| | - Isao Ishii
- Laboratory of Health Chemistry, Showa Pharmaceutical University, Tokyo 194-8543, Japan;
| | - Philip K. Moore
- Department of Pharmacology, National University of Singapore, Singapore 119228, Singapore;
| | - Madhav Bhatia
- Inflammation Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand;
| | - A. Mark Richards
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8140, New Zealand; (L.J.E.); (E.M.T.); (A.P.P.); (C.F.); (A.M.R.)
- Cardiovascular Research Institute, National University of Singapore, Singapore 119228, Singapore
| | - Vicky A. Cameron
- Christchurch Heart Institute, Department of Medicine, University of Otago, Christchurch 8140, New Zealand; (L.J.E.); (E.M.T.); (A.P.P.); (C.F.); (A.M.R.)
| |
Collapse
|
20
|
Hydrogen Sulfide as a Potential Alternative for the Treatment of Myocardial Fibrosis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:4105382. [PMID: 32064023 PMCID: PMC6998763 DOI: 10.1155/2020/4105382] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 12/10/2019] [Indexed: 12/13/2022]
Abstract
Harmful, stressful conditions or events in the cardiovascular system result in cellular damage, inflammation, and fibrosis. Currently, there is no targeted therapy for myocardial fibrosis, which is highly associated with a large number of cardiovascular diseases and can lead to fatal heart failure. Hydrogen sulfide (H2S) is an endogenous gasotransmitter similar to nitric oxide and carbon monoxide. H2S is involved in the suppression of oxidative stress, inflammation, and cellular death in the cardiovascular system. The level of H2S in the body can be boosted by stimulating its synthesis or supplying it exogenously with a simple H2S donor with a rapid- or slow-releasing mode, an organosulfur compound, or a hybrid with known drugs (e.g., aspirin). Hypertension, myocardial infarction, and inflammation are exaggerated when H2S is reduced. In addition, the exogenous delivery of H2S mitigates myocardial fibrosis caused by various pathological conditions, such as a myocardial infarct, hypertension, diabetes, or excessive β-adrenergic stimulation, via its involvement in a variety of signaling pathways. Numerous experimental findings suggest that H2S may work as a potential alternative for the management of myocardial fibrosis. In this review, the antifibrosis role of H2S is briefly addressed in order to gain insight into the development of novel strategies for the treatment of myocardial fibrosis.
Collapse
|
21
|
Nin DS, Idres SB, Song ZJ, Moore PK, Deng LW. Biological Effects of Morpholin-4-Ium 4 Methoxyphenyl (Morpholino) Phosphinodithioate and Other Phosphorothioate-Based Hydrogen Sulfide Donors. Antioxid Redox Signal 2020; 32:145-158. [PMID: 31642346 DOI: 10.1089/ars.2019.7896] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Significance: Hydrogen sulfide (H2S) is regarded as the third gasotransmitter along with nitric oxide and carbon monoxide. Extensive studies have demonstrated a variety of biological roles for H2S in neurophysiology, cardiovascular disease, endocrine regulation, and other physiological and pathological processes. Recent Advances: Novel H2S donors have proved useful in understanding the biological functions of H2S, with morpholin-4-ium 4 methoxyphenyl (morpholino) phosphinodithioate (GYY4137) being one of the most common pharmacological tools used. One advantage of GYY4137 over sulfide salts is its ability to release H2S in a slow and sustained manner akin to endogenous H2S production, rather than the delivery of H2S as a single concentrated burst. Critical Issues: Here, we summarize recent progress made in the characterization of the biological activities and pharmacological effects of GYY4137 in a range of in vitro and in vivo systems. Recent developments in the structural modification of GYY4137 to generate new compounds and their biological effects are also discussed. Future Directions: Slow-releasing H2S donor, GYY4137, and other phosphorothioate-based H2S donors are potent tools to study the biological functions of H2S. Despite recent progress, more work needs to be performed on these new compounds to unravel the mechanisms behind H2S release and pace of its discharge, as well as to define the effects of by-products of donors after H2S liberation. This will not only lead to better in-depth understanding of the biological effects of H2S but will also shed light on the future development of a new class of therapeutic agents with potential to treat a wide range of human diseases.
Collapse
Affiliation(s)
- Dawn Sijin Nin
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Shabana Binte Idres
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhi Jian Song
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Philip K Moore
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Lih-Wen Deng
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,National University Cancer Institute, National University Health System, Singapore, Singapore
| |
Collapse
|
22
|
Majumder S, Ren L, Pushpakumar S, Sen U. Hydrogen sulphide mitigates homocysteine-induced apoptosis and matrix remodelling in mesangial cells through Akt/FOXO1 signalling cascade. Cell Signal 2019; 61:66-77. [PMID: 31085234 PMCID: PMC6561819 DOI: 10.1016/j.cellsig.2019.05.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/28/2019] [Accepted: 05/02/2019] [Indexed: 12/17/2022]
Abstract
Cellular damage and accumulation of extracellular matrix (ECM) protein in the glomerulo-interstitial space are the signatures of chronic kidney disease (CKD). Hyperhomocysteinemia (HHcy), a high level of homocysteine (Hcy) is associated with CKD and further contributes to kidney damage. Despite a large number of studies, the signalling mechanism of Hcy-mediated cellular damage and ECM remodelling in kidney remains inconclusive. Hcy metabolizes to produce hydrogen sulphide (H2S), and a number of studies have shown that H2S mitigates the adverse effect of HHcy in a variety of diseases involving several signalling molecules, including forkhead box O (FOXO) protein. FOXO is a group of transcription factor that includes FOXO1, which plays important roles in cell growth and proliferation. On the other hand, a cell survival factor, Akt regulates FOXO under normal condition. However, the involvement of Akt/FOXO1 pathway in Hcy-induced mesangial cell damage remains elusive, and whether H2S plays any protective roles has yet to be clearly defined. We treated mouse mesangial cells with or without H2S donor, GYY4137 and FOXO1 inhibitor, AS1842856 in HHcy condition and determined the involvement of Akt/FOXO1 signalling cascades. Our results indicated that Hcy inactivated Akt and activated FOXO1 by dephosphorylating both the signalling molecules and induced FOXO1 nuclear translocation followed by activation of the FOXO1 transcription factor. These led to the induction of cellular apoptosis and synthesis of excessive ECM protein, in part, due to increased ROS production, loss of mitochondrial membrane potential (ΔΨm), reduction in intracellular ATP concentration, increased MMP-2, -9, -14 mRNA and protein expression, and Col I, IV and fibronectin protein expression. Interestingly, GYY4137 or AS1842856 treatment prevented these changes by modulating Akt/FOXO1 axis in HHcy. We conclude that GYY4137 and/or AS1842856 mitigates HHcy induced mesangial cell damage and ECM remodelling by regulating Akt/FOXO1 pathway.
Collapse
Affiliation(s)
- Suravi Majumder
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Lu Ren
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, United States of America
| | - Sathnur Pushpakumar
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States of America
| | - Utpal Sen
- Department of Physiology, University of Louisville School of Medicine, Louisville, KY, United States of America.
| |
Collapse
|
23
|
Merz T, Lukaschewski B, Wigger D, Rupprecht A, Wepler M, Gröger M, Hartmann C, Whiteman M, Szabo C, Wang R, Waller C, Radermacher P, McCook O. Interaction of the hydrogen sulfide system with the oxytocin system in the injured mouse heart. Intensive Care Med Exp 2018; 6:41. [PMID: 30341744 PMCID: PMC6195501 DOI: 10.1186/s40635-018-0207-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/07/2018] [Indexed: 02/08/2023] Open
Abstract
Background Both the hydrogen sulfide/cystathionine-γ-lyase (H2S/CSE) and oxytocin/oxytocin receptor (OT/OTR) systems have been reported to be cardioprotective. H2S can stimulate OT release, thereby affecting blood volume and pressure regulation. Systemic hyper-inflammation after blunt chest trauma is enhanced in cigarette smoke (CS)-exposed CSE−/− mice compared to wildtype (WT). CS increases myometrial OTR expression, but to this point, no data are available on the effects CS exposure on the cardiac OT/OTR system. Since a contusion of the thorax (Txt) can cause myocardial injury, the aim of this post hoc study was to investigate the effects of CSE−/− and exogenous administration of GYY4137 (a slow release H2S releasing compound) on OTR expression in the heart, after acute on chronic disease, of CS exposed mice undergoing Txt. Methods This study is a post hoc analysis of material obtained in wild type (WT) homozygous CSE−/− mice after 2-3 weeks of CS exposure and subsequent anesthesia, blast wave-induced TxT, and surgical instrumentation for mechanical ventilation (MV) and hemodynamic monitoring. CSE−/− animals received a 50 μg/g GYY4137-bolus after TxT. After 4h of MV, animals were exsanguinated and organs were harvested. The heart was cut transversally, formalin-fixed, and paraffin-embedded. Immunohistochemistry for OTR, arginine-vasopressin-receptor (AVPR), and vascular endothelial growth factor (VEGF) was performed with naïve animals as native controls. Results CSE−/− was associated with hypertension and lower blood glucose levels, partially and significantly restored by GYY4137 treatment, respectively. Myocardial OTR expression was reduced upon injury, and this was aggravated in CSE−/−. Exogenous H2S administration restored myocardial protein expression to WT levels. Conclusions This study suggests that cardiac CSE regulates cardiac OTR expression, and this effect might play a role in the regulation of cardiovascular function.
Collapse
Affiliation(s)
- Tamara Merz
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany.
| | - Britta Lukaschewski
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany
| | - Daniela Wigger
- Clinic for Psychsomatic Medicine and Psychotherapy, University Medical Center, Ulm, Germany
| | - Aileen Rupprecht
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany
| | - Martin Wepler
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany.,Department of Anesthesiology, University Medical Center, Ulm, Germany
| | - Michael Gröger
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany
| | - Clair Hartmann
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany.,Department of Anesthesiology, University Medical Center, Ulm, Germany
| | - Matthew Whiteman
- University of Exeter Medical School, St. Luke's Campus, Exeter, England, UK
| | - Csaba Szabo
- Chair of Pharmacology, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland.,Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rui Wang
- Department of Biology, Laurentian University, Sudbury, ON, Canada
| | - Christiane Waller
- Clinic for Psychsomatic Medicine and Psychotherapy, University Medical Center, Ulm, Germany.,Department of Psychosomatic Medicine and Psychotherapy, Paracelsus Medical University, Nuremberg General Hospital, Nuremberg, Germany
| | - Peter Radermacher
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany
| | - Oscar McCook
- Institute of Anesthesiological Pathophysiology and Process Engineering, University Medical School, Helmholtzstrasse 8-1, 89081, Ulm, Germany
| |
Collapse
|
24
|
An Update on Hydrogen Sulfide and Nitric Oxide Interactions in the Cardiovascular System. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4579140. [PMID: 30271527 PMCID: PMC6151216 DOI: 10.1155/2018/4579140] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 07/25/2018] [Indexed: 01/19/2023]
Abstract
Hydrogen sulfide (H2S) and nitric oxide (NO) are now recognized as important regulators in the cardiovascular system, although they were historically considered as toxic gases. As gaseous transmitters, H2S and NO share a wide range of physical properties and physiological functions: they penetrate into the membrane freely; they are endogenously produced by special enzymes, they stimulate endothelial cell angiogenesis, they regulate vascular tone, they protect against heart injury, and they regulate target protein activity via posttranslational modification. Growing evidence has determined that these two gases are not independent regulators but have substantial overlapping pathophysiological functions and signaling transduction pathways. H2S and NO not only affect each other's biosynthesis but also produce novel species through chemical interaction. They play a regulatory role in the cardiovascular system involving similar signaling mechanisms or molecular targets. However, the natural precise mechanism of the interactions between H2S and NO remains unclear. In this review, we discuss the current understanding of individual and interactive regulatory functions of H2S and NO in biosynthesis, angiogenesis, vascular one, cardioprotection, and posttranslational modification, indicating the importance of their cross-talk in the cardiovascular system.
Collapse
|
25
|
Abstract
Hydrogen sulfide (H2S) is a novel signaling molecule most recently found to be of fundamental importance in cellular function as a regulator of apoptosis, inflammation, and perfusion. Mechanisms of endogenous H2S signaling are poorly understood; however, signal transmission is thought to occur via persulfidation at reactive cysteine residues on proteins. Although much has been discovered about how H2S is synthesized in the body, less is known about how it is metabolized. Recent studies have discovered a multitude of different targets for H2S therapy, including those related to protein modification, intracellular signaling, and ion channel depolarization. The most difficult part of studying hydrogen sulfide has been finding a way to accurately and reproducibly measure it. The purpose of this review is to: elaborate on the biosynthesis and catabolism of H2S in the human body, review current knowledge of the mechanisms of action of this gas in relation to ischemic injury, define strategies for physiological measurement of H2S in biological systems, and review potential novel therapies that use H2S for treatment.
Collapse
|
26
|
Hofmann F. A concise discussion of the regulatory role of cGMP kinase I in cardiac physiology and pathology. Basic Res Cardiol 2018; 113:31. [PMID: 29934662 DOI: 10.1007/s00395-018-0690-1] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 05/18/2018] [Accepted: 06/13/2018] [Indexed: 12/25/2022]
Abstract
The underlying cause of cardiac hypertrophy, fibrosis, and heart failure has been investigated in great detail using different mouse models. These studies indicated that cGMP and cGMP-dependent protein kinase type I (cGKI) may ameliorate these negative phenotypes in the adult heart. Recently, evidence has been published that cardiac mitochondrial BKCa channels are a target for cGKI and that activation of mitoBKCa channels may cause some of the positive effects of conditioning in ischemia/reperfusion injury. It will be pointed out that most studies could not present convincing evidence that it is the cGMP level and the activity cGKI in specific cardiac cells that reduces hypertrophy or heart failure. However, anti-fibrotic compounds stimulating nitric oxide-sensitive guanylyl cyclase may be an upcoming therapy for abnormal cardiac remodeling.
Collapse
Affiliation(s)
- Franz Hofmann
- Institut für Pharmakologie und Toxikologie, TU München, Biedersteiner Str. 29, 80802, Munich, Germany.
| |
Collapse
|
27
|
Qiu Y, Wu Y, Meng M, Luo M, Zhao H, Sun H, Gao S. GYY4137 protects against myocardial ischemia/reperfusion injury via activation of the PHLPP-1/Akt/Nrf2 signaling pathway in diabetic mice. J Surg Res 2018; 225:29-39. [PMID: 29605032 DOI: 10.1016/j.jss.2017.12.030] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Revised: 11/21/2017] [Accepted: 12/19/2017] [Indexed: 12/31/2022]
Abstract
BACKGROUND This study explores the protective effects of a hydrogen sulfide donor, morpholin-4-ium 4-methoxyphenyl-morpholino-phosphinodithioate (GYY4137), in the hearts of diabetic mice that had been subjected to myocardial ischemia/reperfusion injury. Diabetes impairs the Akt pathway, in which the Akt protein is dephosphorylated and inactivated by PH domain leucine-rich repeat protein phosphatase-1 (PHLPP-1). However, the function of PHLPP-1 and molecular mechanism that underlies the cardiac protection exerted by GYY4137 remains unknown. METHODS Diabetic or nondiabetic mice were subjected to 45 min of coronary artery occlusion followed by 2 h of reperfusion. H9c2 cells were cultured with normal or high glucose and then subjected to 3 h of hypoxia followed by 6 h of reoxygenation. Pretreatment with GYY4137 was performed in a randomized manner before ischemia/reperfusion or hypoxia/reoxygenation. The infarct size, cardiomyocyte apoptosis, and oxidative stress were measured. Western blotting was conducted to elucidate the protective mechanism. RESULTS Diabetic mice or H9c2 cells exposed to high glucose displayed a larger infarct size, more severe cardiomyocyte apoptosis, lower cell viability, and increased oxidative stress, which were associated with increased levels of PHLPP-1 and reduced levels of p-Akt and nuclear factor-erythroid-2-related factor 2 (Nrf2) protein expression. These changes were prevented/reversed by GYYG4137 pretreatment. At the cellular level, PHLPP-1 siRNA attenuated cellular injury, and this was associated with increased p-Akt and nuclear Nrf2 protein, whereas the decrement of Akt phosphorylation induced by LY294002 augmented cellular injury and decreased nuclear Nrf2. CONCLUSIONS GYY4137 activates the PHLPP-1/Akt/Nrf2 pathway to protect against diabetic myocardial ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Yun Qiu
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Yichen Wu
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Min Meng
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Man Luo
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Hongmei Zhao
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Hong Sun
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China
| | - Sumin Gao
- Department of Emergency Medicine, Huai'an First People's Hospital, Nanjing Medical University, Huai'an, Jiangsu Province, China.
| |
Collapse
|
28
|
Murphy PB, Bihari A, Parry NG, Ball I, Leslie K, Vogt K, Lawendy AR. Carbon monoxide and hydrogen sulphide reduce reperfusion injury in abdominal compartment syndrome. J Surg Res 2017; 222:17-25. [PMID: 29273369 DOI: 10.1016/j.jss.2017.09.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 08/28/2017] [Accepted: 09/27/2017] [Indexed: 01/13/2023]
Abstract
BACKGROUND Carbon monoxide (CO)- and hydrogen sulphide-releasing molecules (CORM-3 and GYY4137, respectively) have been shown to be potent antioxidant and antiinflammatory agents at the tissue and systemic level. We hypothesized that both CORM-3 and GYY4137 would reduce the significant organ dysfunction associated with abdominal compartment syndrome (ACS). MATERIAL AND METHODS Randomized trial was conducted where ACS was maintained for 2 hours in 27 rats using an abdominal plaster cast and intraperitoneal CO2 insufflation at 20 mmHg. Three experimental groups underwent ACS and received an experimental molecule at the time of decompression: inactive CORM-3, active CORM-3, and GYY4137, whereas three groups underwent no ACS to serve as a sham. Sinusoidal perfusion, inflammatory response and cell death were quantified in exteriorized livers. Respiratory, liver, and renal dysfunction was assessed biochemically. RESULTS Hepatocellular death and the number of activated leukocytes within postsinusoidal venules were significantly increased in rats with ACS (16-fold increase, 17-fold leukocyte activation, respectively, P < 0.05). Administration of CORM-3 or GYY4137 resulted in a significant decrease of both parameters (P = 0.03 and P = 0.009). ACS resulted in an increase in markers of renal and liver injury; CORM-3 or GYY4137 partially restored levels to those seen in sham animals. Myeloperoxidase was significantly elevated in the ACS group in lung, liver, and small intestine (P = 0.0002, P = 0.01, and P = 0.08, respectively). CORM-3 treatment, but not GYY4137, was able to completely block the response (65 ± 11 U/ml and 92 ± 18 U/ml, respectively versus 110 ± 10U/ml in the ACS group, lung tissue). CONCLUSIONS We have demonstrated the effect of two molecules, CO and hydrogen sulphide, on tempering the reperfusion-associated metabolic and organ derangements in ACS. CORM-3 demonstrated a greater effect than GYY4137 and was able to restore most of the measured parameters to levels comparable to sham.
Collapse
Affiliation(s)
- Patrick B Murphy
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada.
| | - Aurelia Bihari
- Division of Orthopedic Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Neil G Parry
- Divisions of General Surgery and Critical Care, Department of Surgery, Schulich School of Medicine and Dentistry, Trauma Program, London Health Sciences Centre & Divisions of General Surgery and Critical Care Medicine, Western University, London, Ontario, Canada
| | - Ian Ball
- Division of Critical Care, Schulich School of Medicine and Dentistry, Trauma Program, London Health Sciences Centre & Divisions of General Surgery and Critical Care Medicine, Western University, London, Ontario, Canada
| | - Ken Leslie
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Kelly Vogt
- Division of General Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| | - Abdel-Rahman Lawendy
- Division of Orthopedic Surgery, Department of Surgery, Schulich School of Medicine and Dentistry, Western University, London, Ontario, Canada
| |
Collapse
|
29
|
Szabo C, Papapetropoulos A. International Union of Basic and Clinical Pharmacology. CII: Pharmacological Modulation of H 2S Levels: H 2S Donors and H 2S Biosynthesis Inhibitors. Pharmacol Rev 2017; 69:497-564. [PMID: 28978633 PMCID: PMC5629631 DOI: 10.1124/pr.117.014050] [Citation(s) in RCA: 304] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Over the last decade, hydrogen sulfide (H2S) has emerged as an important endogenous gasotransmitter in mammalian cells and tissues. Similar to the previously characterized gasotransmitters nitric oxide and carbon monoxide, H2S is produced by various enzymatic reactions and regulates a host of physiologic and pathophysiological processes in various cells and tissues. H2S levels are decreased in a number of conditions (e.g., diabetes mellitus, ischemia, and aging) and are increased in other states (e.g., inflammation, critical illness, and cancer). Over the last decades, multiple approaches have been identified for the therapeutic exploitation of H2S, either based on H2S donation or inhibition of H2S biosynthesis. H2S donation can be achieved through the inhalation of H2S gas and/or the parenteral or enteral administration of so-called fast-releasing H2S donors (salts of H2S such as NaHS and Na2S) or slow-releasing H2S donors (GYY4137 being the prototypical compound used in hundreds of studies in vitro and in vivo). Recent work also identifies various donors with regulated H2S release profiles, including oxidant-triggered donors, pH-dependent donors, esterase-activated donors, and organelle-targeted (e.g., mitochondrial) compounds. There are also approaches where existing, clinically approved drugs of various classes (e.g., nonsteroidal anti-inflammatories) are coupled with H2S-donating groups (the most advanced compound in clinical trials is ATB-346, an H2S-donating derivative of the non-steroidal anti-inflammatory compound naproxen). For pharmacological inhibition of H2S synthesis, there are now several small molecule compounds targeting each of the three H2S-producing enzymes cystathionine-β-synthase (CBS), cystathionine-γ-lyase, and 3-mercaptopyruvate sulfurtransferase. Although many of these compounds have their limitations (potency, selectivity), these molecules, especially in combination with genetic approaches, can be instrumental for the delineation of the biologic processes involving endogenous H2S production. Moreover, some of these compounds (e.g., cell-permeable prodrugs of the CBS inhibitor aminooxyacetate, or benserazide, a potentially repurposable CBS inhibitor) may serve as starting points for future clinical translation. The present article overviews the currently known H2S donors and H2S biosynthesis inhibitors, delineates their mode of action, and offers examples for their biologic effects and potential therapeutic utility.
Collapse
Affiliation(s)
- Csaba Szabo
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, Texas (C.S.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece (A.P.); and Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Andreas Papapetropoulos
- Department of Anesthesiology, The University of Texas Medical Branch, Galveston, Texas (C.S.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Zografou, Greece (A.P.); and Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| |
Collapse
|
30
|
Yuan S, Shen X, Kevil CG. Beyond a Gasotransmitter: Hydrogen Sulfide and Polysulfide in Cardiovascular Health and Immune Response. Antioxid Redox Signal 2017; 27:634-653. [PMID: 28398086 PMCID: PMC5576200 DOI: 10.1089/ars.2017.7096] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE Hydrogen sulfide (H2S) metabolism leads to the formation of oxidized sulfide species, including polysulfide, persulfide, and others. Evidence is emerging that many biological effects of H2S may indeed be due to polysulfide and persulfide activation of signaling pathways and reactivity with discrete small molecules. Recent Advances: Exogenous oxidized sulfide species, including polysulfides, are more reactive than H2S with a wide range of molecules. Importantly, endogenous polysulfide and persulfide formation has been reported to occur via transsulfuration enzymes, cystathionine γ-lyase (CSE) and cystathionine β-synthase (CBS). CRITICAL ISSUES In light of the recent understanding of oxidized sulfide metabolite formation and reactivity, comparatively few studies have been reported comparing cellular biological and in vivo effects of H2S donors versus polysulfide and persulfide donors. Likewise, it is equally unclear when, how, and to what extent persulfide and polysulfide formation occurs in vivo under pathophysiological conditions. FUTURE DIRECTIONS Additional studies regarding persulfide and polysulfide formation and molecular reactions are needed in nearly all aspects of biology to better understand how sulfide metabolites contribute to key chemical biology reactions involved in cardiovascular health and immune responses. Antioxid. Redox Signal. 27, 634-653.
Collapse
Affiliation(s)
- Shuai Yuan
- 1 Department of Cell Biology and Anatomy, LSU Health Sciences Center Shreveport , Shreveport, Louisiana
| | - Xinggui Shen
- 2 Department of Pathology and Translational Pathobiology, LSU Health Sciences Center Shreveport , Shreveport, Louisiana
| | - Christopher G Kevil
- 2 Department of Pathology and Translational Pathobiology, LSU Health Sciences Center Shreveport , Shreveport, Louisiana
| |
Collapse
|
31
|
Martinez EC, Lilyanna S, Wang P, Vardy LA, Jiang X, Armugam A, Jeyaseelan K, Richards AM. MicroRNA-31 promotes adverse cardiac remodeling and dysfunction in ischemic heart disease. J Mol Cell Cardiol 2017; 112:27-39. [PMID: 28865712 DOI: 10.1016/j.yjmcc.2017.08.013] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 08/08/2017] [Accepted: 08/29/2017] [Indexed: 12/12/2022]
Abstract
RATIONALE Myocardial infarction (MI) triggers a dynamic microRNA response with the potential of yielding therapeutic targets. OBJECTIVE We aimed to identify novel aberrantly expressed cardiac microRNAs post-MI with potential roles in adverse remodeling in a rat model, and to provide post-ischemic therapeutic inhibition of a candidate pathological microRNA in vivo. METHODS AND RESULTS Following microRNA array profiling in rat hearts 2 and 14days post-MI, we identified a time-dependent up-regulation of miR-31 compared to sham-operated rats. A progressive increase of miR-31 (up to 91.4±11.3 fold) was detected in the infarcted myocardium by quantitative real-time PCR. Following target prediction analysis, reporter gene assays confirmed that miR-31 targets the 3´UTR of cardiac troponin-T (Tnnt2), E2F transcription factor 6 (E2f6), mineralocorticoid receptor (Nr3c2) and metalloproteinase inhibitor 4 (Timp4) mRNAs. In vitro, hypoxia and oxidative stress up-regulated miR-31 and suppressed target genes in cardiac cell cultures, whereas LNA-based oligonucleotide inhibition of miR-31 (miR-31i) reversed its repressive effect on target mRNAs. Therapeutic post-ischemic administration of miR-31i in rats silenced cardiac miR-31 and enhanced expression of target genes, while preserving cardiac structure and function at 2 and 4weeks post-MI. Left ventricular ejection fraction (EF) improved by 10% (from day 2 to 30 post-MI) in miR-31i-treated rats, whereas controls receiving scrambled LNA inhibitor or placebo incurred a 17% deterioration in EF. miR-31i decreased end-diastolic pressure and infarct size; attenuated interstitial fibrosis in the remote myocardium and enhanced cardiac output. CONCLUSION miR-31 induction after MI is deleterious to cardiac function while its therapeutic inhibition in vivo ameliorates cardiac dysfunction and prevents the development of post-ischemic adverse remodeling.
Collapse
Affiliation(s)
- Eliana C Martinez
- Cardiovascular Research Institute, National University Health System, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Interdisciplinary Stem Cell Institute, Department of Pediatrics, Division of Cardiology, University of Miami Miller School of Medicine, Miami, FL, USA.
| | - Shera Lilyanna
- Cardiovascular Research Institute, National University Health System, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Peipei Wang
- Cardiovascular Research Institute, National University Health System, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Leah A Vardy
- A*STAR Institute of Medical Biology, Singapore; Department of Biological Sciences, Nanyang Technological University, Singapore
| | - Xiaofei Jiang
- Cardiovascular Research Institute, National University Health System, Singapore
| | - Arunmozhiarasi Armugam
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kandiah Jeyaseelan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Victoria, Australia
| | - Arthur Mark Richards
- Cardiovascular Research Institute, National University Health System, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Cardiac Department, National University Health System, Singapore; Christchurch Heart Institute, University of Otago, Christchurch, New Zealand
| |
Collapse
|
32
|
Sun X, Wang W, Dai J, Jin S, Huang J, Guo C, Wang C, Pang L, Wang Y. A Long-Term and Slow-Releasing Hydrogen Sulfide Donor Protects against Myocardial Ischemia/Reperfusion Injury. Sci Rep 2017; 7:3541. [PMID: 28615705 PMCID: PMC5471203 DOI: 10.1038/s41598-017-03941-0] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 05/05/2017] [Indexed: 01/25/2023] Open
Abstract
Hydrogen sulfide (H2S) has been recognized as an important gasotransmitter exerting various physiological effects, especially in the cardiovascular system. Herein we investigated the cardioprotective effects of a novel long-term and slow-releasing H2S donor, DATS-MSN, using in vivo myocardial ischemia/reperfusion (I/R) models and in vitro hypoxia/reoxygenation cardiomyocyte models. Unlike the instant-releasing pattern of sodium hydrosulphide (NaHS), the release of H2S from DATS-MSN was quite slow and continuous both in the cell culture medium and in rat plasma (elevated H2S concentrations during 24 h and 72 h reperfusion). Correspondingly, DATS-MSN demonstrated superior cardioprotective effects over NaHS in I/R models, which were associated with greater survival rates, reduced CK-MB and troponin I levels, decreased cardiomyocyte apoptosis index, increased antioxidant enzyme activities, inhibited myocardial inflammation, greater reduction in the infarct area and preserved cardiac ejection fraction. Some of these effects of DATS-MSN were also found to be superior to classic slow-releasing H2S donor, GYY4137. In in vitro experiments, cardiomyocytes injury was also found to be relived with the use of DATS-MSN compared to NaHS after the hypoxia/reoxygenation processes. The present work provides a novel long-term and slow-releasing H2S donor and an insight into how the release patterns of H2S donors affect its physiological functionality.
Collapse
Affiliation(s)
- Xiaotian Sun
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, 200040, China.
| | - Wenshuo Wang
- Department of Cardiac Surgery, Zhongshan Hospital of Fudan University and Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Jing Dai
- Department of Cadres Health Care, Third Hospital of Shijiazhuang, Shijiazhuang, 050011, China
| | - Sheng Jin
- Department of Physiology, Hebei Medical University, Shijiazhuang, 050017, China
| | - Jiechun Huang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, 200040, China
| | - Changfa Guo
- Department of Cardiac Surgery, Zhongshan Hospital of Fudan University and Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Chunsheng Wang
- Department of Cardiac Surgery, Zhongshan Hospital of Fudan University and Shanghai Institute of Cardiovascular Diseases, Shanghai, 200032, China
| | - Liewen Pang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, 200040, China.
| | - Yiqing Wang
- Department of Cardiothoracic Surgery, Huashan Hospital of Fudan University, Shanghai, 200040, China.
| |
Collapse
|
33
|
Garcia NA, Moncayo-Arlandi J, Vazquez A, Genovés P, Calvo CJ, Millet J, Martí N, Aguado C, Knecht E, Valiente-Alandi I, Montero JA, Díez-Juan A, Sepúlveda P. Hydrogen Sulfide Improves Cardiomyocyte Function in a Cardiac Arrest Model. Ann Transplant 2017; 22:285-295. [PMID: 28484204 PMCID: PMC6248014 DOI: 10.12659/aot.901410] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Background Cardioplegic arrest is a common procedure for many types of cardiac surgery, and different formulations have been proposed to enhance its cardio-protective effect. Hydrogen sulfide is an important signaling molecule that has cardio-protective properties. We therefore studied the cardio-protective effect of hydrogen sulfide in cardiac cell culture and its potential therapeutic use in combination with cardioplegia formulations. Materials/Methods We added hydrogen sulfide donor GYY4137 to HL-1 cells to study its protective effect in nutrient starved conditions. In addition, we tested the potential use of GYY4137 when it is added into two different cardioplegia formulations: Cardi-Braun® solution and del Nido solution in an ex vivo Langendorff perfused rat hearts model. Results We observed that eight-hour pre-treatment with GYY4137 significantly suppressed apoptosis in nutrient-starved HL-1 cells (28% less compared to untreated cells; p<0.05), maintained ATP content, and reduced protein synthesis. In ex vivo experiments, Cardi-Braun® and del Nido cardioplegia solutions supplemented with GYY4137 significantly reduced the pro-apoptotic protein caspase-3 content and preserved ATP content. Furthermore, GYY4137 supplemented cardioplegia solutions decreased the S-(5-adenosyl)-L-methionine/S-(adenosyl)-L-homocysteine ratio, reducing the oxidative stress in cardiac tissue. Finally, heart beating analysis revealed the preservation of the inter-beat interval and the heart rate in del Nido cardioplegia solution supplemented with GYY4137. Conclusions GYY4137 preconditioning preserved energetic state during starved conditions, attenuating the cardiomyocytes apoptosis in vitro. The addition of GYY4137 to cardioplegia solutions prevented apoptosis, ATP consumption, and oxidative stress in perfused rat hearts, restoring its electrophysiological status after cardiac arrest. These findings suggested that GYY4137 sulfide donor may improve the cardioplegia solution performance during cardiac surgery.
Collapse
Affiliation(s)
- Nahuel Aquiles Garcia
- Mixed Unit for Cardiovascular Repair, Institute of Sanitary Research La Fe-Príncipe Felipe Research Center, Valencia, Spain
| | - Javier Moncayo-Arlandi
- Mixed Unit for Cardiovascular Repair, Institute of Sanitary Research La Fe-Príncipe Felipe Research Center, Valencia, Spain.,Cardiovascular Genetics Center, Institute for Biomedical investigation Girona, Girona, Spain
| | - Alejandro Vazquez
- Mixed Unit for Cardiovascular Repair, Institute of Sanitary Research La Fe-Príncipe Felipe Research Center, Valencia, Spain
| | - Patricia Genovés
- Mixed Unit for Cardiovascular Repair, Institute of Sanitary Research La Fe-Príncipe Felipe Research Center, Valencia, Spain
| | - Conrado J Calvo
- BIOITACA, Cardiovascular Protection Cluster, Universitat Politécnica de Valencia, Valencia, Spain
| | - José Millet
- BIOITACA, Cardiovascular Protection Cluster, Universitat Politécnica de Valencia, Valencia, Spain
| | - Nuria Martí
- Príncipe Felipe Research Center, Valencia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER) ISCIII, Valencia, Spain
| | - Carmen Aguado
- Príncipe Felipe Research Center, Valencia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER) ISCIII, Valencia, Spain
| | - Erwin Knecht
- Príncipe Felipe Research Center, Valencia, Spain.,Center for Biomedical Research on Rare Diseases (CIBERER) ISCIII, Valencia, Spain
| | - Iñigo Valiente-Alandi
- The Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - José A Montero
- Mixed Unit for Cardiovascular Repair, Institute of Sanitary Research La Fe-Príncipe Felipe Research Center, Valencia, Spain
| | | | - Pilar Sepúlveda
- Mixed Unit for Cardiovascular Repair, Institute of Sanitary Research La Fe-Príncipe Felipe Research Center, Valencia, Spain
| |
Collapse
|
34
|
Kanagy NL, Szabo C, Papapetropoulos A. Vascular biology of hydrogen sulfide. Am J Physiol Cell Physiol 2017; 312:C537-C549. [PMID: 28148499 PMCID: PMC5451519 DOI: 10.1152/ajpcell.00329.2016] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/24/2017] [Accepted: 01/27/2017] [Indexed: 12/23/2022]
Abstract
Hydrogen sulfide (H2S) is a ubiquitous signaling molecule with important functions in many mammalian organs and systems. Observations in the 1990s ascribed physiological actions to H2S in the nervous system, proposing that this gasotransmitter acts as a neuromodulator. Soon after that, the vasodilating properties of H2S were demonstrated. In the past decade, H2S was shown to exert a multitude of physiological effects in the vessel wall. H2S is produced by vascular cells and exhibits antioxidant, antiapoptotic, anti-inflammatory, and vasoactive properties. In this concise review, we have focused on the impact of H2S on vascular structure and function with an emphasis on angiogenesis, vascular tone, vascular permeability and atherosclerosis. H2S reduces arterial blood pressure, limits atheromatous plaque formation, and promotes vascularization of ischemic tissues. Although the beneficial properties of H2S are well established, mechanistic insights into the molecular pathways implicated in disease prevention and treatment remain largely unexplored. Unraveling the targets and downstream effectors of H2S in the vessel wall in the context of disease will aid in translation of preclinical observations. In addition, acute regulation of H2S production is still poorly understood and additional work delineating the pathways regulating the enzymes that produce H2S will allow pharmacological manipulation of this pathway. As the field continues to grow, we expect that H2S-related compounds will find their way into clinical trials for diseases affecting the blood vessels.
Collapse
Affiliation(s)
- Nancy L Kanagy
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece; and
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
35
|
Huang P, Shen Z, Yu W, Huang Y, Tang C, Du J, Jin H. Hydrogen Sulfide Inhibits High-Salt Diet-Induced Myocardial Oxidative Stress and Myocardial Hypertrophy in Dahl Rats. Front Pharmacol 2017; 8:128. [PMID: 28360857 PMCID: PMC5352693 DOI: 10.3389/fphar.2017.00128] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 03/01/2017] [Indexed: 12/27/2022] Open
Abstract
The study aimed to examine the protective effect of hydrogen sulfide (H2S) on high-salt-induced oxidative stress and myocardial hypertrophy in salt-sensitive (Dahl) rats. Thirty male Dahl rats and 40 SD rats were included in the study. They were randomly divided into Dahl control (Dahl + NS), Dahl high salt (Dahl + HS), Dahl + HS + NaHS, SD + NS, SD + HS, SD + HS + NaHS, and SD + HS + hydroxylamine (HA). Rats in Dahl + NS and SD + NS groups were given chow with 0.5% NaCl and 0.9% normal saline intraperitoneally daily. Myocardial structure, α-myosin heavy chain (α-MHC) and β-myosin heavy chain (β-MHC) expressions were determined. Endogenous myocardial H2S pathway and oxidative stress in myocardial tissues were tested. Myocardial H2S pathway was downregulated with myocardial hypertrophy featured by increased heart weight/body weight and cardiomyocytes cross-sectional area, decreased α-MHC and increased β-MHC expressions in Dahl rats with high-salt diet (all P < 0.01), and oxidative stress in myocardial tissues was significantly activated, demonstrated by the increased contents of hydroxyl radical, malondialdehyde and oxidized glutathione and decreased total antioxidant capacity, carbon monoxide, catalase, glutathione, glutathione peroxidase, superoxide dismutase (SOD) activities and decreased SOD1 and SOD2 protein expressions (P < 0.05, P < 0.01). However, H2S reduced myocardial hypertrophy with decreased heart weight/body weight and cardiomyocytes cross-sectional area, increased α-MHC, decreased β-MHC expressions and inhibited oxidative stress in myocardial tissues of Dahl rats with high-salt diet. However, no significant difference was found in H2S pathway, myocardial structure, α-MHC and β-MHC protein and oxidative status in myocardial tissues among SD + NS, SD + HS, and SD + HS + NaHS groups. HA, an inhibitor of cystathionine β-synthase, inhibited myocardial H2S pathway (P < 0.01), and stimulated myocardial hypertrophy and oxidative stress in SD rats with high-salt diet. Hence, H2S inhibited myocardial hypertrophy in high salt-stimulated Dahl rats in association with the enhancement of antioxidant capacity, thereby inhibiting oxidative stress in myocardial tissues.
Collapse
Affiliation(s)
- Pan Huang
- Department of Pediatrics, Peking University First Hospital Beijing, China
| | - Zhizhou Shen
- Department of Pediatrics, Peking University First Hospital Beijing, China
| | - Wen Yu
- Department of Pediatrics, Peking University First Hospital Beijing, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital Beijing, China
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Centre Beijing, China
| | - Junbao Du
- Department of Pediatrics, Peking University First HospitalBeijing, China; Key Laboratory of Molecular Cardiology, Ministry of Education, Peking UniversityBeijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital Beijing, China
| |
Collapse
|
36
|
Grambow E, Leppin C, Leppin K, Kundt G, Klar E, Frank M, Vollmar B. The effects of hydrogen sulfide on platelet-leukocyte aggregation and microvascular thrombolysis. Platelets 2016; 28:509-517. [PMID: 27819526 DOI: 10.1080/09537104.2016.1235693] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The volatile transmitter hydrogen sulfide (H2S) is known for its various functions in vascular biology. This study evaluates the effect of the H2S-donor GYY4137 (GYY) on thrombus stability and microvascular thrombolysis. Human whole blood served for all in vitro studies and was analyzed in a resting state, after stimulation with thrombin-receptor activating peptide (TRAP) and after incubation with 10 or 30 mM GYY or its vehicle DMSO following TRAP-activation, respectively. As a marker for thrombus stability, platelet-leukocyte aggregation was assessed using flow cytometry after staining of human whole blood against CD62P and CD45, respectively. Furthermore, morphology and quantity of platelet-leukocyte aggregation were studied by means of scanning electron microscopy (scanning EM). Therefore, platelets were stained for CD62P followed by immuno gold labeling. In vivo, the dorsal skinfold chamber preparation was performed for light/dye induction of thrombi in arterioles and venules using intravital fluorescence microscopy. Thrombolysis was assessed 10 and 22 h after thrombus induction and treatment with the vehicle, GYY, or recombinant tissue plasminogen activator (rtPA). Flow cytometry revealed an increase of CD62P/CD45 positive aggregates after TRAP stimulation of human whole blood, which was significantly reduced by preincubation with 30 mM GYY. Scanning EM additionally showed a reduced platelet-leukocyte aggregation and a decreased leukocyte count within the aggregates after preincubation with GYY compared to TRAP stimulation alone. Further on, morphological signs of platelet activation were found markedly reduced upon treatment with GYY. In mice, both GYY and rtPA significantly accelerated arteriolar and venular thrombolysis compared to the vehicle control. In conclusion, GYY impairs thrombus stability by reducing platelet-leukocyte aggregation and thereby facilitates endogenous thrombolysis.
Collapse
Affiliation(s)
- Eberhard Grambow
- a Institute for Experimental Surgery, Rostock University Medical Center , Rostock , Germany.,b Department of General , Thoracic, Vascular and Transplantation Surgery, Rostock University Medical Center , Rostock , Germany
| | - Christian Leppin
- a Institute for Experimental Surgery, Rostock University Medical Center , Rostock , Germany
| | - Katja Leppin
- a Institute for Experimental Surgery, Rostock University Medical Center , Rostock , Germany
| | - Günther Kundt
- c Institute for Biostatistics and Informatics in Medicine and Aging Research, Rostock University Medical Center , Rostock , Germany
| | - Ernst Klar
- b Department of General , Thoracic, Vascular and Transplantation Surgery, Rostock University Medical Center , Rostock , Germany
| | - Marcus Frank
- d Medical Biology and Electron Microscopy Centre, Rostock University Medical Center , Rostock , Germany
| | - Brigitte Vollmar
- a Institute for Experimental Surgery, Rostock University Medical Center , Rostock , Germany
| |
Collapse
|
37
|
Chatzianastasiou A, Bibli SI, Andreadou I, Efentakis P, Kaludercic N, Wood ME, Whiteman M, Di Lisa F, Daiber A, Manolopoulos VG, Szabó C, Papapetropoulos A. Cardioprotection by H2S Donors: Nitric Oxide-Dependent and ‑Independent Mechanisms. J Pharmacol Exp Ther 2016; 358:431-40. [PMID: 27342567 PMCID: PMC6047225 DOI: 10.1124/jpet.116.235119] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 06/21/2016] [Indexed: 12/27/2022] Open
Abstract
Hydrogen sulfide (H2S) is a signaling molecule with protective effects in the cardiovascular system. To harness the therapeutic potential of H2S, a number of donors have been developed. The present study compares the cardioprotective actions of representative H2S donors from different classes and studies their mechanisms of action in myocardial injury in vitro and in vivo. Exposure of cardiomyocytes to H2O2 led to significant cytotoxicity, which was inhibited by sodium sulfide (Na2S), thiovaline (TV), GYY4137 [morpholin-4-ium 4 methoxyphenyl(morpholino) phosphinodithioate], and AP39 [(10-oxo-10-(4-(3-thioxo-3H-1,2-dithiol5yl)phenoxy)decyl) triphenylphospho-nium bromide]. Inhibition of nitric oxide (NO) synthesis prevented the cytoprotective effects of Na2S and TV, but not GYY4137 and AP39, against H2O2-induced cardiomyocyte injury. Mice subjected to left anterior descending coronary ligation were protected from ischemia-reperfusion injury by the H2S donors tested. Inhibition of nitric oxide synthase (NOS) in vivo blocked only the beneficial effect of Na2S. Moreover, Na2S, but not AP39, administration enhanced the phosphorylation of endothelial NOS and vasodilator-associated phosphoprotein. Both Na2S and AP39 reduced infarct size in mice lacking cyclophilin-D (CypD), a modulator of the mitochondrial permeability transition pore (PTP). Nevertheless, only AP39 displayed a direct effect on mitochondria by increasing the mitochondrial Ca(2+) retention capacity, which is evidence of decreased propensity to undergo permeability transition. We conclude that although all the H2S donors we tested limited infarct size, the pathways involved were not conserved. Na2S had no direct effects on PTP opening, and its action was nitric oxide dependent. In contrast, the cardioprotection exhibited by AP39 could result from a direct inhibitory effect on PTP acting at a site different than CypD.
Collapse
Affiliation(s)
- Athanasia Chatzianastasiou
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Sofia-Iris Bibli
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Ioanna Andreadou
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Panagiotis Efentakis
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Nina Kaludercic
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Mark E Wood
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Matthew Whiteman
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Fabio Di Lisa
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Andreas Daiber
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Vangelis G Manolopoulos
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Csaba Szabó
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| | - Andreas Papapetropoulos
- George P. Livanos and Marianthi Simou Laboratories, First Department of Pulmonary and Critical Care Medicine, Evangelismos Hospital, Faculty of Medicine, National and Kapodistrian University of Athens, Athens, Greece (A.C., A.P.); Laboratory of Pharmacology, Democritus University of Thrace Medical School, Alexandroupolis, Greece (A.C., V.G.M.); Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece (S.-I.B., I.A., P.E., A.P.); Neuroscience Institute, CNR, Italy (N.K., F.D.L.); Biosciences, College of Life and Environmental Sciences, University of Exeter, Exeter, United Kingdom (M.E.W.); University of Exeter Medical School, Exeter, United Kingdom (M.W.); Department of Biomedical Sciences, University of Padova, Padova, Italy (F.D.L.); Center of Cardiology and Center for Thrombosis and Hemostasis, Medical Center of the Johannes Gutenberg University, Mainz, Germany (A.D.); Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas (C.S.); Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece (A.P.)
| |
Collapse
|
38
|
Chen S, Bu D, Ma Y, Zhu J, Sun L, Zuo S, Ma J, Li T, Chen Z, Zheng Y, Wang X, Pan Y, Wang P, Liu Y. GYY4137 ameliorates intestinal barrier injury in a mouse model of endotoxemia. Biochem Pharmacol 2016; 118:59-67. [PMID: 27553476 DOI: 10.1016/j.bcp.2016.08.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 08/15/2016] [Indexed: 01/13/2023]
Abstract
Intestinal barrier injury has been reported to play a vital role in the pathogenesis of endotoxemia. This study aimed to investigate the protective effect of GYY4137, a newly synthesized H2S donor, on the intestinal barrier function in the context of endotoxemia both in vitro and in vivo. Caco-2 (a widely used human colon cancer cell line in the study of intestinal epithelial barrier function) monolayers incubated with lipopolysaccharide (LPS) or TNF-α/IFN-γ and a mouse model of endotoxemia were used in this study. The results suggested that GYY4137 significantly attenuated LPS or TNF-α/IFN-γ induced increased Caco-2 monolayer permeability. The decreased expression of TJ (tight junction) proteins induced by LPS and the altered localization of TJs induced by TNF-α/IFN-γ was significantly inhibited by GYY4137; similar results were obtained in vivo. Besides, GYY4137 promoted the clinical score and histological score of mice with endotoxemia. Increased level of TNF-α/IFN-γ in the plasma and increased apoptosis in colon epithelial cells was also attenuated by GYY4137 in mice with endotoxemia. This study indicates that GYY4137 preserves the intestinal barrier function in the context of endotoxemia via multipathways and throws light on the development of potential therapeutic approaches for endotoxemia.
Collapse
Affiliation(s)
- Shanwen Chen
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Dingfang Bu
- Central Laboratory, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Yuanyuan Ma
- Animal Experiment Center, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Jing Zhu
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Lie Sun
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Shuai Zuo
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Ju Ma
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Tengyu Li
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Zeyang Chen
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Youwen Zheng
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Xin Wang
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Yisheng Pan
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China
| | - Pengyuan Wang
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China.
| | - Yucun Liu
- Division of General Surgery, Peking University First Hospital, Peking University, 8 Xi ShiKu Street, Beijing 100034, People's Republic of China.
| |
Collapse
|
39
|
Liu HD, Zhang AJ, Xu JJ, Chen Y, Zhu YC. H2S protects against fatal myelosuppression by promoting the generation of megakaryocytes/platelets. J Hematol Oncol 2016; 9:13. [PMID: 26912146 PMCID: PMC4766725 DOI: 10.1186/s13045-016-0244-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/17/2016] [Indexed: 12/13/2022] Open
Abstract
Background Our previous pilot studies aimed to examine the role of hydrogen sulfide (H2S) in the generation of endothelial progenitor cells led to an unexpected result, i.e., H2S promoted the differentiation of certain hematopoietic stem/progenitor cells in the bone marrow. This gave rise to an idea that H2S might promote hematopoiesis. Methods To test this idea, a mice model of myelosuppression and cultured fetal liver cells were used to examine the role of H2S in hematopoiesis. Results H2S promoted the generation of megakaryocytes, increased platelet levels, ameliorate entorrhagia, and improved survival. These H2S effects were blocked in both in vivo and in vitro models with thrombopoietin (TPO) receptor knockout mice (c-mpl−/− mice). In contrast, H2S promoted megakaryocytes/platelets generation in both in vivo and in vitro models with TPO knockout mice (TPO−/− mice). Conclusions H2S is a novel promoter for megakaryopoiesis by acting on the TPO receptors but not TPO to generate megakaryocytes/platelets. Electronic supplementary material The online version of this article (doi:10.1186/s13045-016-0244-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Huan-Di Liu
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, 138 Yi Xue Yuan Road, Shanghai, 200032, China.,Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine in Henan Province, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, China
| | - Ai-Jie Zhang
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Jing-Jing Xu
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, 138 Yi Xue Yuan Road, Shanghai, 200032, China.,Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying Chen
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, 138 Yi Xue Yuan Road, Shanghai, 200032, China
| | - Yi-Chun Zhu
- Shanghai Key Laboratory of Bioactive Small Molecules and Research Center on Aging and Medicine, Department of Physiology and Pathophysiology, Fudan University Shanghai Medical College, 138 Yi Xue Yuan Road, Shanghai, 200032, China.
| |
Collapse
|