1
|
Mallardi D, Chimienti G, Maqoud F, Orlando A, Drago S, Malerba E, De Virgilio C, Akbarali HI, Russo F. The Dual Role of Exogenous Hydrogen Sulfide (H 2S) in Intestinal Barrier Mitochondrial Function: Insights into Cytoprotection and Cytotoxicity Under Non-Stressed Conditions. Antioxidants (Basel) 2025; 14:384. [PMID: 40298652 PMCID: PMC12024010 DOI: 10.3390/antiox14040384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/30/2025] Open
Abstract
Hydrogen sulfide (H2S) is a critical gasotransmitter that plays a dual role in physiological and pathological processes, particularly in the gastrointestinal tract. While physiological levels of H2S exert cytoprotective effects, excessive concentrations can lead to toxicity, oxidative stress, and inflammation. The aim of this study was to investigate the dose-dependent effects of exogenous H2S on mitochondrial functions and biogenesis in intestinal epithelial cells under non-stressed conditions. Using a Caco-2 monolayer model, we evaluated the impact of sodium hydrosulfide (NaHS) at concentrations ranging from 1 × 10-7 M to 5 × 10-3 M on mitochondrial metabolism, redox balance, antioxidant defense, inflammatory responses, autophagy/mitophagy, and apoptosis. Our results demonstrated a biphasic response: low-to-moderate H2S concentrations (1 × 10-7 M-1.5 × 10-3 M) enhance mitochondrial biogenesis through PGC-1α activation, upregulating TFAM and COX-4 expression, and increasing the mtDNA copy number. In contrast, higher concentrations (2 × 10-3-5 × 10-3 M) impair mitochondrial function, induce oxidative stress, and promote apoptosis. These effects are associated with elevated reactive oxygen species (ROS) production, dysregulation of antioxidant enzymes, and COX-2-mediated inflammation. H2S-induced autophagy/mitophagy is a protective mechanism at intermediate concentrations but fails to mitigate mitochondrial damage at toxic levels. This study underscores the delicate balance between the cytoprotective and cytotoxic effects of exogenous H2S in intestinal cells, helping to develop new therapeutic approaches for gastrointestinal disorders.
Collapse
Affiliation(s)
- Domenica Mallardi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (D.M.); (F.M.); (A.O.); (S.D.); (E.M.)
| | - Guglielmina Chimienti
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (G.C.); (C.D.V.)
| | - Fatima Maqoud
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (D.M.); (F.M.); (A.O.); (S.D.); (E.M.)
| | - Antonella Orlando
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (D.M.); (F.M.); (A.O.); (S.D.); (E.M.)
| | - Simona Drago
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (D.M.); (F.M.); (A.O.); (S.D.); (E.M.)
| | - Eleonora Malerba
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (D.M.); (F.M.); (A.O.); (S.D.); (E.M.)
| | - Caterina De Virgilio
- Department of Biosciences, Biotechnologies and Environment, University of Bari Aldo Moro, 70125 Bari, Italy; (G.C.); (C.D.V.)
| | - Hamid I. Akbarali
- Department of Pharmacology and Toxicology, School of Medicine, Virginia Commonwealth University, Richmond, VA 23298, USA;
| | - Francesco Russo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (D.M.); (F.M.); (A.O.); (S.D.); (E.M.)
| |
Collapse
|
2
|
Wang YW, Chu T, Wang XL, Fan YQ, Cao L, Chen YH, Zhu YW, Liu HX, Ji XY, Wu DD. The role of cystathionine β-synthase in cancer. Cell Signal 2024; 124:111406. [PMID: 39270916 DOI: 10.1016/j.cellsig.2024.111406] [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: 07/12/2024] [Revised: 08/28/2024] [Accepted: 09/09/2024] [Indexed: 09/15/2024]
Abstract
Cystathionine β-synthase (CBS) occupies a key position as the initiating and rate-limiting enzyme in the sulfur transfer pathway and plays a vital role in health and disease. CBS is responsible for regulating the metabolism of cysteine, the precursor of glutathione (GSH), an important antioxidant in the body. Additionally, CBS is one of the three enzymes that produce hydrogen sulfide (H2S) in mammals through a variety of mechanisms. The dysregulation of CBS expression in cancer cells affects H2S production through direct or indirect pathways, thereby influencing cancer growth and metastasis by inducing angiogenesis, facilitating proliferation, migration, and invasion, modulating cellular energy metabolism, promoting cell cycle progression, and inhibiting apoptosis. It is noteworthy that CBS expression exhibits complex changes in different cancer models. In this paper, we focus on the CBS synthesis and metabolism, tissue distribution, potential mechanisms influencing tumor growth, and relevant signaling pathways. We also discuss the impact of pharmacological CBS inhibitors and silencing CBS in preclinical cancer models, supporting their potential as targeted cancer therapies.
Collapse
Affiliation(s)
- Yan-Wen Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Ti Chu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Xue-Li Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Yong-Qi Fan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Lei Cao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Yu-Hang Chen
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Yi-Wen Zhu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China
| | - Hong-Xia Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan 450064, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, School of Stomatology, Henan University, Kaifeng, Henan 475004, China; Department of Stomatology, Huaihe Hospital of Henan University, School of Stomatology, Henan University, Kaifeng, Henan 475004, China.
| |
Collapse
|
3
|
Wu J, Zhao Q, Chen S, Xu H, Zhang R, Cai D, Gao Y, Peng W, Chen X, Yuan S, Li D, Li G, Nan A. NSUN4-mediated m5C modification of circERI3 promotes lung cancer development by altering mitochondrial energy metabolism. Cancer Lett 2024; 605:217266. [PMID: 39332589 DOI: 10.1016/j.canlet.2024.217266] [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: 05/18/2024] [Revised: 09/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024]
Abstract
As a highly important methylation modification, the 5-methyladenosine (m5C) modification can profoundly affect RNAs by regulating their transcription, structure and stability. With the continuous development of high-throughput technology, differentially expressed circular RNAs (circRNAs) have been increasingly discovered, and circRNAs play unique roles in tumorigenesis and development. However, the regulatory mechanism of the m5C modification of circRNAs has not yet been revealed. In this study, circERI3, which is highly expressed in lung cancer tissue and significantly correlated with the clinical progression of lung cancer, was initially identified through differential expression profiling of circRNAs. A combined m5C microarray analysis revealed that circERI3 contains the m5C modification and that the NSUN4-mediated m5C modification of circERI3 can increase its nuclear export. The important function of circERI3 in promoting lung cancer progression in vitro and in vivo was clarified. Moreover, we elucidated the novel mechanism by which circERI3 targets DNA binding protein 1 (DDB1), regulates its ubiquitination, enhances its stability, and in turn promotes the transcription of peroxisome proliferator-activated receptor γ coactivator 1α (PGC-1α) through DDB1 to affect mitochondrial function and energy metabolism, which ultimately promotes the development of lung cancer. This study not only revealed the reasons for the abnormal distribution of circERI3 in lung cancer tissues from the perspective of methylation and clarified the important role of circERI3 in lung cancer progression but also described a novel mechanism by which circERI3 promotes lung cancer development through mitochondrial energy metabolism, providing new insights for the study of circRNAs in lung cancer.
Collapse
Affiliation(s)
- Jiaxi Wu
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Qingyun Zhao
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Sixian Chen
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Haotian Xu
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Ruirui Zhang
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Dunyu Cai
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Yihong Gao
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Wenyi Peng
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Xingcai Chen
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Shengyi Yuan
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Deqing Li
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China
| | - Gang Li
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| | - Aruo Nan
- School of Public Health, Guangxi Medical University, Nanning 530021, China; Guangxi Key Laboratory of Environment and Health Research, Guangxi Medical University, Nanning, 530021, China.
| |
Collapse
|
4
|
Zhou J, Zheng X, Xi C, Tang X, Jiang Y, Xie M, Fu X. Cr(VI) induced hepatocyte apoptosis through the CTH/H 2S/Drp1 signaling pathway. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 950:175332. [PMID: 39117219 DOI: 10.1016/j.scitotenv.2024.175332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/04/2024] [Accepted: 08/04/2024] [Indexed: 08/10/2024]
Abstract
Hexavalent chromium [Cr(VI)] is a highly hazardous heavy metal with multiple toxic effects. Occupational studies indicate that its accumulation in humans can lead to liver damage. However, the exact mechanism underlying Cr(VI)-induced hepatotoxicity remains unknown. In this study, we explored the role of CTH/H2S/Drp1 pathway in Cr(VI)-induced oxidative stress, mitochondrial dysfunction, apoptosis, and liver injury. Our data showed that Cr(VI) triggered apoptosis, accompanied by H2S reduction, reactive oxygen species (ROS) accumulation, and mitochondrial dysfunction in both AML12 cells and mouse livers. Moreover, Cr(VI) reduced cystathionine γ-lyase (CTH) and dynamin related protein 1 (Drp1) S-sulfhydration levels, and elevated Drp1 phosphorylation levels at Serine 616, which promoted Drp1 mitochondrial translocation and Drp1-voltage-dependent anion channel 1 (VDAC1) interactions, ultimately leading to mitochondria-dependent apoptosis. Elevated hydrogen sulfide (H2S) levels eliminated Drp1 phosphorylation at Serine 616 by increasing Drp1 S-sulfhydration, thereby preventing Cr(VI)-induced Drp1-VDAC1 interaction and hepatotoxicity. These findings indicated that Cr(VI) induced mitochondrial apoptosis and hepatotoxicity by inhibiting CTH/H2S/Drp1 pathway and that targeting either CTH/H2S pathway or Drp1 S-sulfhydration could serve as a potential therapy for Cr(VI)-induced liver injury.
Collapse
Affiliation(s)
- Jie Zhou
- School of Medicine, Yichun University,576 XueFu Road, Yuanzhou District, Yichun 336000, PR China.
| | - Xin Zheng
- School of Medicine, Yichun University,576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Chen Xi
- Pharmaceutical Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, PR China
| | - Xinyi Tang
- School of Medicine, Yichun University,576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Yinjie Jiang
- School of Medicine, Yichun University,576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Minjuan Xie
- School of Medicine, Yichun University,576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| | - Xiaoyi Fu
- School of Medicine, Yichun University,576 XueFu Road, Yuanzhou District, Yichun 336000, PR China
| |
Collapse
|
5
|
Mahmoud DSE, Kamel MA, El-Sayed IET, Binsuwaidan R, Elmongy EI, Razzaq MK, Abd Eldaim MA, Ahmed ESAM, Shaker SA. Astaxanthin ameliorated isoproterenol induced myocardial infarction via improving the mitochondrial function and antioxidant activity in rats. J Biochem Mol Toxicol 2024; 38:e23804. [PMID: 39132813 DOI: 10.1002/jbt.23804] [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: 02/16/2024] [Revised: 05/20/2024] [Accepted: 07/24/2024] [Indexed: 08/13/2024]
Abstract
The present study evaluated the cardioprotective effect of astaxanthin (ASX) against isoproterenol (ISO) induced myocardial infarction in rats via the pathway of mitochondrial biogenesis as the possible molecular target of astaxanthin. The control group was injected with normal physiological saline subcutaneously for 2 days. The second group was injected with ISO at a dose of 85 mg/kg bwt subcutaneously for 2 days. The third, fourth and fifth groups were supplemented with ASX at doses of 10, 20, 30 mg/kg bwt, respectively daily by oral gavage for 21 days then injected with ISO dose of 85 mg/kg bwt subcutaneously for 2 successive days. Isoproterenol administration in rats elevated the activities of Creatine kinase-MB (CK-MB), aspartate transaminase (AST), lactate dehydrogenase (LDH), and other serum cardiac biomarkers Troponin-I activities, oxidative stress biomarkers, malondialdehyde(MDA), Nuclear factor-kappa B (NF-KB), while it decreased Peroxisome proliferator-activated receptor-gamma coactivator (PGC-1α), Nuclear factor erythroid-2-related factor 2 (Nfe212), mitochondrial transcriptional factor A (mt TFA), mitochondrial DNA copy number and glutathione system parameters. However, Astaxanthin decreased the activities of serum AST, LDH, CK-MB, and Troponin I that elevated by ISO. In addition, it increased glutathione peroxidase and reductase activities, total glutathione and reduced GSH content, and GSH/GSSG ratio, mtDNA copy number, PGC-1α expression and Tfam expression that improved mitochondrial biogenesis while it decreased GSSG and MDA contents and NF-KB level in the cardiac tissues. This study indicated that astaxanthin relieved isoproterenol induced myocardial infarction via scavenging free radicals and reducing oxidative damage and apoptosis in cardiac tissue.
Collapse
Affiliation(s)
| | - Maher A Kamel
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| | | | - Reem Binsuwaidan
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia
| | - Elshaymaa I Elmongy
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Helwan University, Cairo, Egypt
| | - Mohand Kareem Razzaq
- Department of Biochemistry, College of Medicine, University of Sumer, Thi-Qar, Iraq
| | - Mabrouk Attia Abd Eldaim
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Menoufia University, Shibin El-Kom, Egypt
| | | | - Sara A Shaker
- Department of Biochemistry, Medical Research Institute, Alexandria University, Alexandria, Egypt
| |
Collapse
|
6
|
Slade L, Deane CS, Szewczyk NJ, Etheridge T, Whiteman M. Hydrogen sulfide supplementation as a potential treatment for primary mitochondrial diseases. Pharmacol Res 2024; 203:107180. [PMID: 38599468 DOI: 10.1016/j.phrs.2024.107180] [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: 01/19/2024] [Revised: 04/06/2024] [Accepted: 04/06/2024] [Indexed: 04/12/2024]
Abstract
Primary mitochondrial diseases (PMD) are amongst the most common inborn errors of metabolism causing fatal outcomes within the first decade of life. With marked heterogeneity in both inheritance patterns and physiological manifestations, these conditions present distinct challenges for targeted drug therapy, where effective therapeutic countermeasures remain elusive within the clinic. Hydrogen sulfide (H2S)-based therapeutics may offer a new option for patient treatment, having been proposed as a conserved mitochondrial substrate and post-translational regulator across species, displaying therapeutic effects in age-related mitochondrial dysfunction and neurodegenerative models of mitochondrial disease. H2S can stimulate mitochondrial respiration at sites downstream of common PMD-defective subunits, augmenting energy production, mitochondrial function and reducing cell death. Here, we highlight the primary signalling mechanisms of H2S in mitochondria relevant for PMD and outline key cytoprotective proteins/pathways amenable to post-translational restoration via H2S-mediated persulfidation. The mechanisms proposed here, combined with the advent of potent mitochondria-targeted sulfide delivery molecules, could provide a framework for H2S as a countermeasure for PMD disease progression.
Collapse
Affiliation(s)
- Luke Slade
- University of Exeter Medical School, University of Exeter, St. Luke's Campus, Exeter EX1 2LU, UK; Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany
| | - Colleen S Deane
- Human Development & Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, UK
| | - Nathaniel J Szewczyk
- Medical Research Council Versus Arthritis Centre for Musculoskeletal Ageing Research, Royal Derby Hospital, University of Nottingham, Derby DE22 3DT, United Kingdom; Ohio Musculoskeletal and Neurologic Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, Ohio 45701, Greece
| | - Timothy Etheridge
- Public Health and Sport Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter EX1 2LU, United Kingdom.
| | - Matthew Whiteman
- University of Exeter Medical School, University of Exeter, St. Luke's Campus, Exeter EX1 2LU, UK.
| |
Collapse
|
7
|
Gao W, Liu YF, Zhang YX, Wang Y, Jin YQ, Yuan H, Liang XY, Ji XY, Jiang QY, Wu DD. The potential role of hydrogen sulfide in cancer cell apoptosis. Cell Death Discov 2024; 10:114. [PMID: 38448410 PMCID: PMC10917771 DOI: 10.1038/s41420-024-01868-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 02/05/2024] [Accepted: 02/14/2024] [Indexed: 03/08/2024] Open
Abstract
For a long time, hydrogen sulfide (H2S) has been considered a toxic compound, but recent studies have found that H2S is the third gaseous signaling molecule which plays a vital role in physiological and pathological conditions. Currently, a large number of studies have shown that H2S mediates apoptosis through multiple signaling pathways to participate in cancer occurrence and development, for example, PI3K/Akt/mTOR and MAPK signaling pathways. Therefore, the regulation of the production and metabolism of H2S to mediate the apoptotic process of cancer cells may improve the effectiveness of cancer treatment. In this review, the role and mechanism of H2S in cancer cell apoptosis in mammals are summarized.
Collapse
Affiliation(s)
- Wei Gao
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Ya-Fang Liu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yan-Xia Zhang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yan Wang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yu-Qing Jin
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Hang Yuan
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xiao-Yi Liang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Xin-Ying Ji
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
- Faculty of Basic Medical Subjects, Shu-Qing Medical College of Zhengzhou, Zhengzhou, Henan, 450064, China.
| | - Qi-Ying Jiang
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
| | - Dong-Dong Wu
- Henan International Joint Laboratory for Nuclear Protein Regulation, School of Basic Medical Sciences, Henan University, Kaifeng, Henan, 475004, China.
- School of Stomatology, Henan University, Kaifeng, Henan, 475004, China.
- Department of Stomatology, Huaihe Hospital of Henan University, Kaifeng, Henan, 475000, China.
| |
Collapse
|
8
|
Song Y, Cao H, Zuo C, Gu Z, Huang Y, Miao J, Fu Y, Guo Y, Jiang Y, Wang F. Mitochondrial dysfunction: A fatal blow in depression. Biomed Pharmacother 2023; 167:115652. [PMID: 37801903 DOI: 10.1016/j.biopha.2023.115652] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/08/2023] Open
Abstract
Mitochondria maintain the normal physiological function of nerve cells by producing sufficient cellular energy and performing crucial roles in maintaining the metabolic balance through intracellular Ca2+ homeostasis, oxidative stress, and axonal development. Depression is a prevalent psychiatric disorder with an unclear pathophysiology. Damage to the hippocampal neurons is a key component of the plasticity regulation of synapses and plays a critical role in the mechanism of depression. There is evidence suggesting that mitochondrial dysfunction is associated with synaptic impairment. The maintenance of mitochondrial homeostasis includes quantitative maintenance and quality control of mitochondria. Mitochondrial biogenesis produces new and healthy mitochondria, and mitochondrial dynamics cooperates with mitophagy to remove damaged mitochondria. These processes maintain mitochondrial population stability and exert neuroprotective effects against early depression. In contrast, mitochondrial dysfunction is observed in various brain regions of patients with major depressive disorders. The accumulation of defective mitochondria accelerates cellular nerve dysfunction. In addition, impaired mitochondria aggravate alterations in the brain microenvironment, promoting neuroinflammation and energy depletion, thereby exacerbating the development of depression. This review summarizes the influence of mitochondrial dysfunction and the underlying molecular pathways on the pathogenesis of depression. Additionally, we discuss the maintenance of mitochondrial homeostasis as a potential therapeutic strategy for depression.
Collapse
Affiliation(s)
- Yu Song
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Huan Cao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Chengchao Zuo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Zhongya Gu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yaqi Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Jinfeng Miao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yufeng Fu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yu Guo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yongsheng Jiang
- Cancer Center of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, 430030 Hubei, China.
| | - Furong Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China; Key Laboratory of Vascular Aging (HUST), Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, 430030 Hubei, China.
| |
Collapse
|
9
|
Wang H, Bai Q, Ma G. The biological functions of protein S-sulfhydration in eukaryotes and the ever-increasing understanding of its effects on bacteria. Microbiol Res 2023; 271:127366. [PMID: 36989759 DOI: 10.1016/j.micres.2023.127366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Revised: 02/21/2023] [Accepted: 03/17/2023] [Indexed: 03/29/2023]
Abstract
As a critical endogenous signaling molecule, hydrogen sulfide may induce reversible post-translational modifications on cysteine residues of proteins, generating a persulfide bond known as S-sulfhydration. A systemic overview of the biofunctions of S-sulfhydration will equip us better to characterize its regulatory roles in antioxidant defense, inflammatory response, and cell fate, as well as its pathological mechanisms related to cardiovascular, neurological, and multiple organ diseases, etc. Nevertheless, the understanding of S-sulfhydration is mostly built on mammalian cells and animal models. We subsequently summarized the mediation effects of this specific post-transcriptional modification on physiological processes and virulence in bacteria. The high-sensitivity and high-throughput detection technologies are required for studying the signal transduction mechanism of H2S and protein S-sulfhydration modification. Herein, we reviewed the establishment and development of different approaches to assess S-sulfhydration, including the biotin-switch method, modified biotin-switch method, alkylation-based cysteine-labelled assay, and Tag-switch method. Finally, we discussed the limitations of the impacts of S-sulfhydration in pathogens-host interactions and envisaged the challenges to design drugs and antibiotics targeting the S-sulfhydrated proteins in the host or pathogens.
Collapse
|
10
|
Huang S, Chen X, Pan J, Zhang H, Ke J, Gao L, Yu Chang AC, Zhang J, Zhang H. Hydrogen sulfide alleviates heart failure with preserved ejection fraction in mice by targeting mitochondrial abnormalities via PGC-1α. Nitric Oxide 2023; 136-137:12-23. [PMID: 37182786 DOI: 10.1016/j.niox.2023.05.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 05/03/2023] [Accepted: 05/11/2023] [Indexed: 05/16/2023]
Abstract
AIM Increasing evidence has proposed that mitochondrial abnormalities may be an important factor contributing to the development of heart failure with preserved ejection fraction (HFpEF). Hydrogen sulfide (H2S) has been suggested to play a pivotal role in regulating mitochondrial function. Therefore, the present study was designed to explore the protective effect of H2S on mitochondrial dysfunction in a multifactorial mouse model of HFpEF. METHODS Wild type, 8-week-old, male C57BL/6J mice or cardiomyocyte specific-Cse (Cystathionine γ-lyase, a major H2S-producing enzyme) knockout mice (CSEcko) were given high-fat diet (HFD) and l-NAME (an inhibitor of constitutive nitric oxide synthases) or standardized chow. After 4 weeks, mice were randomly administered with NaHS (a conventional H2S donor), ZLN005 (a potent transcriptional activator of PGC-1α) or vehicle. After additional 4 weeks, echocardiogram and mitochondrial function were evaluated. Expression of PGC-1α, NRF1 and TFAM in cardiomyocytes was assayed by western blot. RESULTS Challenging with HFD and l-NAME in mice not only caused HFpEF but also inhibited the production of endogenous H2S in a time-dependent manner. Meanwhile the expression of PGC-1α and mitochondrial function in cardiomyocytes were impaired. Supplementation with NaHS not only upregulated the expression of PGC-1α, NRF1 and TFAM in cardiomyocytes but also restored mitochondrial function and ultrastructure, conferring an obvious improvement in cardiac diastolic function. In contrast, cardiac deletion of CSE gene aggravated the inhibition of PGC-1α-NRF1-TFAM pathway, mitochondrial abnormalities and diastolic dysfunction. The deleterious effect observed in CSEcko HFpEF mice was partially counteracted by pre-treatment with ZLN005 or supplementation with NaHS. CONCLUSION Our findings have demonstrated that H2S ameliorates left ventricular diastolic dysfunction by restoring mitochondrial abnormalities via upregulating PGC-1α and its downstream targets NRF1 and TFAM, suggesting the therapeutic potential of H2S supplementation in multifactorial HFpEF.
Collapse
Affiliation(s)
- Shuying Huang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xiaonan Chen
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jianan Pan
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hui Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Jiahan Ke
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Lin Gao
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Alex Chia Yu Chang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China; Shanghai Institute of Precision Medicine, Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junfeng Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Huili Zhang
- Department of Cardiology, Shanghai Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| |
Collapse
|
11
|
Advances of H2S in Regulating Neurodegenerative Diseases by Preserving Mitochondria Function. Antioxidants (Basel) 2023; 12:antiox12030652. [PMID: 36978900 PMCID: PMC10044936 DOI: 10.3390/antiox12030652] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 02/22/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Neurotoxicity is induced by different toxic substances, including environmental chemicals, drugs, and pathogenic toxins, resulting in oxidative damage and neurodegeneration in mammals. The nervous system is extremely vulnerable to oxidative stress because of its high oxygen demand. Mitochondria are the main source of ATP production in the brain neuron, and oxidative stress-caused mitochondrial dysfunction is implicated in neurodegenerative diseases. H2S was initially identified as a toxic gas; however, more recently, it has been recognized as a neuromodulator as well as a neuroprotectant. Specifically, it modulates mitochondrial activity, and H2S oxidation in mitochondria produces various reactive sulfur species, thus modifying proteins through sulfhydration. This review focused on highlighting the neuron modulation role of H2S in regulating neurodegenerative diseases through anti-oxidative, anti-inflammatory, anti-apoptotic and S-sulfhydration, and emphasized the importance of H2S as a therapeutic molecule for neurological diseases.
Collapse
|
12
|
Zhao X, Cao Y, Zhao E, Li T, Cong T, Gao Y, Zhang J. The Expression Levels of SARS-CoV-2 Infection-Mediating Molecules Promoted by Interferon-γ and Tumor Necrosis Factor-α Are Downregulated by Hydrogen Sulfide. Int J Mol Sci 2022; 23:13624. [PMID: 36362417 PMCID: PMC9656571 DOI: 10.3390/ijms232113624] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2022] [Revised: 10/28/2022] [Accepted: 11/05/2022] [Indexed: 09/07/2024] Open
Abstract
Autoimmune thyroid diseases (AITDs), which include Hashimoto's thyroiditis (HT) and Graves' disease (GD), have a higher prevalence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in the literature. The effects of AITD-associated cytokines on SARS-CoV-2 infection-mediating molecule levels might be involved in the pathogenesis of susceptibility. We speculated that hydrogen sulfide (H2S) might attenuate this process since H2S has antiviral effects. Using immunohistochemistry, we found that angiotensin-converting enzyme-II (ACE2) expression was higher in the HT group and neuropilin 1 (NRP1) expression was higher in HT and GD groups than in the normal group, while transmembrane protease serine type 2 (TMPRSS2) expression was lower in HT and GD groups. When culturing primary thyrocytes with cytokines or sodium hydrosulfide (NaHS) plus cytokines, we found that ACE2 and NRP1 mRNA levels were upregulated while TMPRSS2 levels were downregulated by interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α). After pretreatment with NaHS in thyrocytes, ACE2 and NRP1 expression were downregulated compared to IFN-γ or TNF-α treatment, and NaHS had no effect on TMPRSS2 expression. Our findings suggested that IFN-γ and TNF-α, which are elevated in AITDs, promoted ACE2 and NRP1 expression and inhibited TMPRSS2 expression. H2S might protect against SARS-CoV-2 infection by downregulating ACE2 and NRP1 levels.
Collapse
Affiliation(s)
- Xue Zhao
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Yedi Cao
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Enmin Zhao
- Department of Otolaryngology, Peking University First Hospital, Beijing 100034, China
| | - Tiancheng Li
- Department of Otolaryngology, Peking University First Hospital, Beijing 100034, China
| | - Tiechuan Cong
- Department of Otolaryngology, Peking University First Hospital, Beijing 100034, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
13
|
Khot M, Sood A, Tryphena KP, Khan S, Srivastava S, Singh SB, Khatri DK. NLRP3 inflammasomes: A potential target to improve mitochondrial biogenesis in Parkinson's disease. Eur J Pharmacol 2022; 934:175300. [PMID: 36167151 DOI: 10.1016/j.ejphar.2022.175300] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/18/2022] [Accepted: 09/21/2022] [Indexed: 11/16/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative condition for which no approved treatment exists to prevent collective neuronal death. There is ample evidence that mitochondrial dysfunction, reactive oxygen species (ROS), and associated caspase activity underlie the pathology observed. Neurons rely on mitochondrial activity since they have such high energy consumption. Therefore, it is not surprising that mitochondrial alterations favour neuronal degeneration. In particular, mitochondrial dysregulation contributes to PD, based on the observation that mitochondrial toxins can cause parkinsonism in humans and animal models. Also, it is known that inflammatory cytokine-mediated neuroinflammation is the key pathogenic mechanism in neuronal loss. In recent years, the research has focussed on mitochondria being the platform for nucleotide-binding oligomerization domain-like receptors 3 (NLRP3) inflammasome activation. Mitochondrial dysfunction and NLRP3 activation are emerging as critical players in inducing and sustaining neuroinflammation. Moreover, mitochondrial-derived ROS and mitochondrial DNA (mtDNA) could serve as the priming signal for forming inflammasome complexes responsible for the activation, maturation, and release of pro-inflammatory cytokines, including interleukin-1(IL-1) and interleukin-18 (IL-18). The current review takes a more comprehensive approach to elucidating the link between mitochondrial dysfunction and aberrant NLRP3 activation in PD. In addition, we focus on some inhibitors of NLRP3 inflammatory pathways to alleviate the progression of PD.
Collapse
Affiliation(s)
- Mayuri Khot
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Anika Sood
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Kamatham Pushpa Tryphena
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Sabiya Khan
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Shashi Bala Singh
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India
| | - Dharmendra Kumar Khatri
- Molecular and Cellular Neuroscience Lab, Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, 500037, India.
| |
Collapse
|
14
|
Persad KL, Lopaschuk GD. Energy Metabolism on Mitochondrial Maturation and Its Effects on Cardiomyocyte Cell Fate. Front Cell Dev Biol 2022; 10:886393. [PMID: 35865630 PMCID: PMC9294643 DOI: 10.3389/fcell.2022.886393] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/20/2022] [Indexed: 12/12/2022] Open
Abstract
Alterations in energy metabolism play a major role in the lineage of cardiomyocytes, such as the dramatic changes that occur in the transition from neonate to newborn. As cardiomyocytes mature, they shift from a primarily glycolytic state to a mitochondrial oxidative metabolic state. Metabolic intermediates and metabolites may have epigenetic and transcriptional roles in controlling cell fate by increasing mitochondrial biogenesis. In the maturing cardiomyocyte, such as in the postnatal heart, fatty acid oxidation increases in conjunction with increased mitochondrial biogenesis driven by the transcriptional coregulator PGC1-α. PGC1-α is necessary for mitochondrial biogenesis in the heart at birth, with deficiencies leading to postnatal cardiomyopathy. While stem cell therapy as a treatment for heart failure requires further investigation, studies suggest that adult stem cells may secrete cardioprotective factors which may regulate cardiomyocyte differentiation and survival. This review will discuss how metabolism influences mitochondrial biogenesis and how mitochondrial biogenesis influences cell fate, particularly in the context of the developing cardiomyocyte. The implications of energy metabolism on stem cell differentiation into cardiomyocytes and how this may be utilized as a therapy against heart failure and cardiovascular disease will also be discussed.
Collapse
|
15
|
Ascenção K, Szabo C. Emerging roles of cystathionine β-synthase in various forms of cancer. Redox Biol 2022; 53:102331. [PMID: 35618601 PMCID: PMC9168780 DOI: 10.1016/j.redox.2022.102331] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 12/12/2022] Open
Abstract
The expression of the reverse transsulfuration enzyme cystathionine-β-synthase (CBS) is markedly increased in many forms of cancer, including colorectal, ovarian, lung, breast and kidney, while in other cancers (liver cancer and glioma) it becomes downregulated. According to the clinical database data in high-CBS-expressor cancers (e.g. colon or ovarian cancer), high CBS expression typically predicts lower survival, while in the low-CBS-expressor cancers (e.g. liver cancer), low CBS expression is associated with lower survival. In the high-CBS expressing tumor cells, CBS, and its product hydrogen sulfide (H2S) serves as a bioenergetic, proliferative, cytoprotective and stemness factor; it also supports angiogenesis and epithelial-to-mesenchymal transition in the cancer microenvironment. The current article reviews the various tumor-cell-supporting roles of the CBS/H2S axis in high-CBS expressor cancers and overviews the anticancer effects of CBS silencing and pharmacological CBS inhibition in various cancer models in vitro and in vivo; it also outlines potential approaches for biomarker identification, to support future targeted cancer therapies based on pharmacological CBS inhibition.
Collapse
|
16
|
Cirino G, Szabo C, Papapetropoulos A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol Rev 2022; 103:31-276. [DOI: 10.1152/physrev.00028.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
H2S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H2S and oxidative posttranscriptional modification of proteins, the effect of H2S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H2S in various cell types and organ systems are overviewed. Finally, the role of H2S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H2S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H2S in the physiological regulation of all organ functions emerges from this review.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece & Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
17
|
Gupta R, Sahu M, Tripathi R, Ambasta RK, Kumar P. Protein S-sulfhydration: Unraveling the prospective of hydrogen sulfide in the brain, vasculature and neurological manifestations. Ageing Res Rev 2022; 76:101579. [PMID: 35124235 DOI: 10.1016/j.arr.2022.101579] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S) and hydrogen polysulfides (H2Sn) are essential regulatory signaling molecules generated by the entire body, including the central nervous system. Researchers have focused on the classical H2S signaling from the past several decades, whereas the last decade has shown the emergence of H2S-induced protein S-sulfhydration signaling as a potential therapeutic approach. Cysteine S-persulfidation is a critical paradigm of post-translational modification in the process of H2S signaling. Additionally, studies have shown the cross-relationship between S-sulfhydration and other cysteine-induced post-translational modifications, namely nitrosylation and carbonylation. In the central nervous system, S-sulfhydration is involved in the cytoprotection through various signaling pathways, viz. inflammatory response, oxidative stress, endoplasmic reticulum stress, atherosclerosis, thrombosis, and angiogenesis. Further, studies have demonstrated H2S-induced S-sulfhydration in regulating different biological processes, such as mitochondrial integrity, calcium homeostasis, blood-brain permeability, cerebral blood flow, and long-term potentiation. Thus, protein S-sulfhydration becomes a crucial regulatory molecule in cerebrovascular and neurodegenerative diseases. Herein, we first described the generation of intracellular H2S followed by the application of H2S in the regulation of cerebral blood flow and blood-brain permeability. Further, we described the involvement of S-sulfhydration in different biological and cellular functions, such as inflammatory response, mitochondrial integrity, calcium imbalance, and oxidative stress. Moreover, we highlighted the importance of S-sulfhydration in cerebrovascular and neurodegenerative diseases.
Collapse
|
18
|
Zhao X, Cao Y, Jin H, Wang X, Zhang L, Zhang Y, Yu Y, Huang Y, Gao Y, Zhang J. Hydrogen Sulfide Promotes Thyroid Hormone Synthesis and Secretion by Upregulating Sirtuin-1. Front Pharmacol 2022; 13:838248. [PMID: 35222046 PMCID: PMC8866871 DOI: 10.3389/fphar.2022.838248] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/17/2022] [Indexed: 11/15/2022] Open
Abstract
Objective: One mechanism of hypothyroidism involves the disruption of thyroid hormone synthesis and secretion by thyrocytes. Hydrogen sulfide (H2S), as a gas signaling molecule, participates in many physiopathologic processes by upregulating sirtuin-1 (SIRT1). The aim of the current study was to explore whether H2S promotes the synthesis and secretion of thyroid hormones by upregulating SIRT1. Methods: Real-time PCR and immunohistochemistry were used to detect the mRNA and protein expression of H2S-generating enzymes in normal human thyroid tissues. Serum H2S concentrations from hypothyroid patients (n = 32) and euthyroid participants (n = 41) were detected by H2S-selective sensors. Thirty-one Sprague–Dawley rats were divided into control group (n = 10), hypothyroid group (induced by MMI, n = 10) and hypothyroid + NaHS group (n = 11), and the FT4, TT4 and TSH levels were assayed. Human primary thyrocytes were incubated with H2S donor sodium hydrosulfide (NaHS) or NaHS plus SIRT1 inhibitor (EX527) in vitro. Thyroid hormone synthesis- and secretion-related proteins [thyroid peroxidase (TPO), sodium iodide transporter (NIS), Pendrin, monocarboxylic acid transporter 8 (MCT8)] were analyzed by real-time PCR and Western blot. Results: H2S levels in serum from hypothyroid patients were decreased compared to those from euthyroid participants (p < .05), and serum H2S levels were positively correlated with FT3, FT4, TT3, and TT4 levels in all subjects (all p < .0001). In vivo, NaHS promoted thyroid function in hypothyroid rats (p < .05). In vitro, H2S was detected in supernatant, and CBS mRNA was higher than CSE and 3-MPST in human primary thyrocytes (p < .05). The protein levels of TPO, NIS, Pendrin and MCT8 were upregulated in a concentration-dependent manner for NaHS in thyrocytes. After blocking SIRT1 with EX527, we found that the increasing levels of TPO, NIS, Pendrin, and MCT8 and TPO activity were downregulated in thyrocytes incubated with NaHS, and FT4 levels in the cell supernatant were also decreased significantly (all p < .05). Conclusion: H2S is mainly generated in thyrocytes by CBS. Serum H2S levels are decreased with hypothyroidism. H2S promotes the synthesis and secretion of thyroid hormones and the expression of related molecules by upregulating SIRT1.
Collapse
Affiliation(s)
- Xue Zhao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Yedi Cao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Xiuli Wang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Lanbo Zhang
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Yang Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Yang Yu
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Youyuan Huang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| | - Ying Gao
- Department of Endocrinology, Peking University First Hospital, Beijing, China
- *Correspondence: Ying Gao,
| | - Junqing Zhang
- Department of Endocrinology, Peking University First Hospital, Beijing, China
| |
Collapse
|
19
|
Elesela S, Lukacs NW. Role of Mitochondria in Viral Infections. Life (Basel) 2021; 11:life11030232. [PMID: 33799853 PMCID: PMC7998235 DOI: 10.3390/life11030232] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/09/2021] [Accepted: 03/10/2021] [Indexed: 12/15/2022] Open
Abstract
Viral diseases account for an increasing proportion of deaths worldwide. Viruses maneuver host cell machinery in an attempt to subvert the intracellular environment favorable for their replication. The mitochondrial network is highly susceptible to physiological and environmental insults, including viral infections. Viruses affect mitochondrial functions and impact mitochondrial metabolism, and innate immune signaling. Resurgence of host-virus interactions in recent literature emphasizes the key role of mitochondria and host metabolism on viral life processes. Mitochondrial dysfunction leads to damage of mitochondria that generate toxic compounds, importantly mitochondrial DNA, inducing systemic toxicity, leading to damage of multiple organs in the body. Mitochondrial dynamics and mitophagy are essential for the maintenance of mitochondrial quality control and homeostasis. Therefore, metabolic antagonists may be essential to gain a better understanding of viral diseases and develop effective antiviral therapeutics. This review briefly discusses how viruses exploit mitochondrial dynamics for virus proliferation and induce associated diseases.
Collapse
Affiliation(s)
- Srikanth Elesela
- Department of Pathology, Michigan Medicine, Ann Arbor, MI 48109, USA
- Correspondence:
| | - Nicholas W. Lukacs
- Mary H. Weiser Food Allergy Center, Michigan Medicine, Ann Arbor, MI 48109, USA;
| |
Collapse
|
20
|
Schiliro M, Bartman CM, Pabelick C. Understanding hydrogen sulfide signaling in neonatal airway disease. Expert Rev Respir Med 2021; 15:351-372. [PMID: 33086886 PMCID: PMC10599633 DOI: 10.1080/17476348.2021.1840981] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/20/2020] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Airway dysfunction leading to chronic lung disease is a common consequence of premature birth and mechanisms responsible for early and progressive airway remodeling are not completely understood. Current therapeutic options are only partially effective in reducing the burden of neonatal airway disease and premature decline of lung function. Gasotransmitter hydrogen sulfide (H2S) has been recently recognized for its therapeutic potential in lung diseases. AREAS COVERED Contradictory to its well-known toxicity at high concentrations, H2S has been characterized to have anti-inflammatory, antioxidant, and antiapoptotic properties at physiological concentrations. In the respiratory system, endogenous H2S production participates in late lung development and exogenous H2S administration has a protective role in a variety of diseases such as acute lung injury and chronic pulmonary hypertension and fibrosis. Literature searches performed using NCBI PubMed without publication date limitations were used to construct this review, which highlights the dichotomous role of H2S in the lung, and explores its promising beneficial effects in lung diseases. EXPERT OPINION The emerging role of H2S in pathways involved in chronic lung disease of prematurity along with its recent use in animal models of BPD highlight H2S as a potential novel candidate in protecting lung function following preterm birth.
Collapse
Affiliation(s)
- Marta Schiliro
- Departments of Anesthesiology, Mayo Clinic, Rochester, MN, USA
| | | | - Christina Pabelick
- Departments of Anesthesiology, Mayo Clinic, Rochester, MN, USA
- Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
21
|
Fox BC, Slade L, Torregrossa R, Pacitti D, Szabo C, Etheridge T, Whiteman M. The mitochondria-targeted hydrogen sulfide donor AP39 improves health and mitochondrial function in a C. elegans primary mitochondrial disease model. J Inherit Metab Dis 2021; 44:367-375. [PMID: 33325042 DOI: 10.1002/jimd.12345] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 12/10/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022]
Abstract
Primary mitochondrial diseases (PMD) are inherited diseases that cause dysfunctional mitochondrial oxidative phosphorylation, leading to diverse multisystem diseases and substantially impaired quality of life. PMD treatment currently comprises symptom management, with an unmet need for therapies targeting the causative mitochondrial defects. Molecules which selective target mitochondria have been proposed as potential treatment options in PMD but have met with limited success. We have previously shown in animal models that mitochondrial dysfunction caused by the disease process could be prevented and/or reversed by selective targeting of the "gasotransmitter" hydrogen sulfide (H2 S) to mitochondria using a novel compound, AP39. Therefore, in this study we investigated whether AP39 could also restore mitochondrial function in PMD models where mitochondrial dysfunction was the cause of the disease pathology using C. elegans. We characterised several PMD mutant C. elegans strains for reduced survival, movement and impaired cellular bioenergetics and treated each with AP39. In animals with widespread electron transport chain deficiency (gfm-1[ok3372]), AP39 (100 nM) restored ATP levels, but had no effect on survival or movement. However, in a complex I mutant (nuo-4[ok2533]), a Leigh syndrome orthologue, AP39 significantly reversed the decline in ATP levels, preserved mitochondrial membrane potential and increased movement and survival. For the first time, this study provides proof-of-principle evidence suggesting that selective targeting of mitochondria with H2 S could represent a novel drug discovery approach to delay, prevent and possibly reverse mitochondrial decline in PMD and related disorders.
Collapse
Affiliation(s)
| | - Luke Slade
- University of Exeter Medical School, Exeter, UK
- College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | | | | | - Csaba Szabo
- Department of Pharmacology, University of Fribourg, Fribourg, Switzerland
| | - Timothy Etheridge
- College of Life and Environmental Sciences, University of Exeter, Exeter, UK
| | | |
Collapse
|
22
|
Szabo C. Hydrogen Sulfide, an Endogenous Stimulator of Mitochondrial Function in Cancer Cells. Cells 2021; 10:cells10020220. [PMID: 33499368 PMCID: PMC7911547 DOI: 10.3390/cells10020220] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Revised: 01/20/2021] [Accepted: 01/21/2021] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2S) has a long history as toxic gas and environmental hazard; inhibition of cytochrome c oxidase (mitochondrial Complex IV) is viewed as a primary mode of its cytotoxic action. However, studies conducted over the last two decades unveiled multiple biological regulatory roles of H2S as an endogenously produced mammalian gaseous transmitter. Cystathionine γ-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST) are currently viewed as the principal mammalian H2S-generating enzymes. In contrast to its inhibitory (toxicological) mitochondrial effects, at lower (physiological) concentrations, H2S serves as a stimulator of electron transport in mammalian mitochondria, by acting as an electron donor—with sulfide:quinone oxidoreductase (SQR) being the immediate electron acceptor. The mitochondrial roles of H2S are significant in various cancer cells, many of which exhibit high expression and partial mitochondrial localization of various H2S producing enzymes. In addition to the stimulation of mitochondrial ATP production, the roles of endogenous H2S in cancer cells include the maintenance of mitochondrial organization (protection against mitochondrial fission) and the maintenance of mitochondrial DNA repair (via the stimulation of the assembly of mitochondrial DNA repair complexes). The current article overviews the state-of-the-art knowledge regarding the mitochondrial functions of endogenously produced H2S in cancer cells.
Collapse
Affiliation(s)
- Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, CH-1700 Fribourg, Switzerland
| |
Collapse
|
23
|
Morris G, Walker AJ, Walder K, Berk M, Marx W, Carvalho AF, Maes M, Puri BK. Increasing Nrf2 Activity as a Treatment Approach in Neuropsychiatry. Mol Neurobiol 2021; 58:2158-2182. [PMID: 33411248 DOI: 10.1007/s12035-020-02212-w] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023]
Abstract
Nuclear factor erythroid 2-related factor 2 (Nrf2) is a transcription factor encoded by NFE2L2. Under oxidative stress, Nrf2 does not undergo its normal cytoplasmic degradation but instead travels to the nucleus, where it binds to a DNA promoter and initiates transcription of anti-oxidative genes. Nrf2 upregulation is associated with increased cellular levels of glutathione disulfide, glutathione peroxidase, glutathione transferases, thioredoxin and thioredoxin reductase. Given its key role in governing the cellular antioxidant response, upregulation of Nrf2 has been suggested as a common therapeutic target in neuropsychiatric illnesses such as major depressive disorder, bipolar disorder and schizophrenia, which are associated with chronic oxidative and nitrosative stress, characterised by elevated levels of reactive oxygen species, nitric oxide and peroxynitrite. These processes lead to extensive lipid peroxidation, protein oxidation and carbonylation, and oxidative damage to nuclear and mitochondrial DNA. Intake of N-acetylcysteine, coenzyme Q10 and melatonin is accompanied by increased Nrf2 activity. N-acetylcysteine intake is associated with improved cerebral mitochondrial function, decreased central oxidative and nitrosative stress, reduced neuroinflammation, alleviation of endoplasmic reticular stress and suppression of the unfolded protein response. Coenzyme Q10, which acts as a superoxide scavenger in neuroglial mitochondria, instigates mitohormesis, ameliorates lipid peroxidation in the inner mitochondrial membrane, activates uncoupling proteins, promotes mitochondrial biogenesis and has positive effects on the plasma membrane redox system. Melatonin, which scavenges mitochondrial free radicals, inhibits mitochondrial nitric oxide synthase, restores mitochondrial calcium homeostasis, deacetylates and activates mitochondrial SIRT3, ameliorates increased permeability of the blood-brain barrier and intestine and counters neuroinflammation and glutamate excitotoxicity.
Collapse
Affiliation(s)
- G Morris
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - A J Walker
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - K Walder
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - M Berk
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia.,CMMR Strategic Research Centre, School of Medicine, Deakin University, Geelong, VIC, Australia.,Orygen, The National Centre of Excellence in Youth Mental Health, The Department of Psychiatry and the Florey Institute for Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, Australia
| | - W Marx
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia
| | - A F Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada.,Centre for Addiction and Mental Health (CAMH), Toronto, ON, Canada
| | - M Maes
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Barwon Health, School of Medicine, Deakin University, Geelong, VIC, Australia.,Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | | |
Collapse
|
24
|
Sun HJ, Wu ZY, Nie XW, Bian JS. The Role of H 2S in the Metabolism of Glucose and Lipids. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1315:51-66. [PMID: 34302688 DOI: 10.1007/978-981-16-0991-6_3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Glucose and lipids are essential elements for maintaining the body's homeostasis, and their dysfunction may participate in the pathologies of various diseases, particularly diabetes, obesity, metabolic syndrome, cardiovascular ailments, and cancers. Among numerous endogenous mediators, the gasotransmitter hydrogen sulfide (H2S) plays a central role in the maintenance of glucose and lipid homeostasis. Current evidence from both pharmacological studies and transgenic animal models suggest a complex relationship between H2S and metabolic dysregulation, especially in diabetes and obesity. This notion is achieved through tissue-specific expressions and actions of H2S on target metabolic and hormone organs including the pancreas, skeletal muscle, livers, and adipose. In this chapter, we will summarize the roles and mechanisms of H2S in several metabolic organs/tissues that are necessary for glucose and lipid metabolic homeostasis. In addition, future research directions and valuable therapeutic avenues around the pharmacological regulation of H2S in glycolipid metabolism disorder will be also discussed.
Collapse
Affiliation(s)
- Hai-Jian Sun
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Zhi-Yuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Xiao-Wei Nie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.,Center of Clinical Research, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, China
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore. .,National University of Singapore (Suzhou) Research Institute, Suzhou, China.
| |
Collapse
|
25
|
Ali A, Wang Y, Wu L, Yang G. Gasotransmitter signaling in energy homeostasis and metabolic disorders. Free Radic Res 2020; 55:83-105. [PMID: 33297784 DOI: 10.1080/10715762.2020.1862827] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Gasotransmitters are small molecules of gases, including nitric oxide (NO), hydrogen sulfide (H2S), and carbon monoxide (CO). These three gasotransmitters can be endogenously produced and regulate a wide range of pathophysiological processes by interacting with specific targets upon diffusion in the biological media. By redox and epigenetic regulation of various physiological functions, NO, H2S, and CO are critical for the maintenance of intracellular energy homeostasis. Accumulated evidence has shown that these three gasotransmitters control ATP generation, mitochondrial biogenesis, glucose metabolism, insulin sensitivity, lipid metabolism, and thermogenesis, etc. Abnormal generation and metabolism of NO, H2S, and/or CO are involved in various abnormal metabolic diseases, including obesity, diabetes, and dyslipidemia. In this review, we summarized the roles of NO, H2S, and CO in the regulation of energy homeostasis as well as their involvements in the metabolism of dysfunction-related diseases. Understanding the interaction among these gasotransmitters and their specific molecular targets are very important for therapeutic applications.
Collapse
Affiliation(s)
- Amr Ali
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Canada.,Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Yuehong Wang
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Canada.,Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - Lingyun Wu
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada.,School of Human Kinetics, Laurentian University, Sudbury, Canada.,Health Science North Research Institute, Sudbury, Canada
| | - Guangdong Yang
- Department of Chemistry and Biochemistry, Laurentian University, Sudbury, Canada.,Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| |
Collapse
|
26
|
Bibli SI, Hu J, Looso M, Weigert A, Ratiu C, Wittig J, Drekolia MK, Tombor L, Randriamboavonjy V, Leisegang MS, Goymann P, Delgado Lagos F, Fisslthaler B, Zukunft S, Kyselova A, Justo AFO, Heidler J, Tsilimigras D, Brandes RP, Dimmeler S, Papapetropoulos A, Knapp S, Offermanns S, Wittig I, Nishimura SL, Sigala F, Fleming I. Mapping the Endothelial Cell S-Sulfhydrome Highlights the Crucial Role of Integrin Sulfhydration in Vascular Function. Circulation 2020; 143:935-948. [PMID: 33307764 DOI: 10.1161/circulationaha.120.051877] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND In vascular endothelial cells, cysteine metabolism by the cystathionine γ lyase (CSE), generates hydrogen sulfide-related sulfane sulfur compounds (H2Sn), that exert their biological actions via cysteine S-sulfhydration of target proteins. This study set out to map the "S-sulfhydrome" (ie, the spectrum of proteins targeted by H2Sn) in human endothelial cells. METHODS Liquid chromatography with tandem mass spectrometry was used to identify S-sulfhydrated cysteines in endothelial cell proteins and β3 integrin intraprotein disulfide bond rearrangement. Functional studies included endothelial cell adhesion, shear stress-induced cell alignment, blood pressure measurements, and flow-induced vasodilatation in endothelial cell-specific CSE knockout mice and in a small collective of patients with endothelial dysfunction. RESULTS Three paired sample sets were compared: (1) native human endothelial cells isolated from plaque-free mesenteric arteries (CSE activity high) and plaque-containing carotid arteries (CSE activity low); (2) cultured human endothelial cells kept under static conditions or exposed to fluid shear stress to decrease CSE expression; and (3) cultured endothelial cells exposed to shear stress to decrease CSE expression and treated with solvent or the slow-releasing H2Sn donor, SG1002. The endothelial cell "S-sulfhydrome" consisted of 3446 individual cysteine residues in 1591 proteins. The most altered family of proteins were the integrins and focusing on β3 integrin in detail we found that S-sulfhydration affected intraprotein disulfide bond formation and was required for the maintenance of an extended-open conformation of the β leg. β3 integrin S-sulfhydration was required for endothelial cell mechanotransduction in vitro as well as flow-induced dilatation in murine mesenteric arteries. In cultured cells, the loss of S-sulfhydration impaired interactions between β3 integrin and Gα13 (guanine nucleotide-binding protein subunit α 13), resulting in the constitutive activation of RhoA (ras homolog family member A) and impaired flow-induced endothelial cell realignment. In humans with atherosclerosis, endothelial function correlated with low H2Sn generation, impaired flow-induced dilatation, and failure to detect β3 integrin S-sulfhydration, all of which were rescued after the administration of an H2Sn supplement. CONCLUSIONS Vascular disease is associated with marked changes in the S-sulfhydration of endothelial cell proteins involved in mediating responses to flow. Short-term H2Sn supplementation improved vascular reactivity in humans highlighting the potential of interfering with this pathway to treat vascular disease.
Collapse
Affiliation(s)
- Sofia-Iris Bibli
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Jiong Hu
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Mario Looso
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.).,Bioinformatics Core Unit (M.L., P.G.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Germany (A.W.)
| | - Corina Ratiu
- Centre for Molecular Medicine, Institute for Cardiovascular Physiology (C.R., M.S.L., R.P.B.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Janina Wittig
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Maria Kyriaki Drekolia
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany
| | - Lukas Tombor
- Institute for Cardiovascular Regeneration (L.T., S.D.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Voahanginirina Randriamboavonjy
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Matthias S Leisegang
- Centre for Molecular Medicine, Institute for Cardiovascular Physiology (C.R., M.S.L., R.P.B.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Philipp Goymann
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.).,Bioinformatics Core Unit (M.L., P.G.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Fredy Delgado Lagos
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Beate Fisslthaler
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Sven Zukunft
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Anastasia Kyselova
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Alberto Fernando Oliveira Justo
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Juliana Heidler
- Functional Proteomics (J.Heidler., I.W.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Diamantis Tsilimigras
- First Propedeutic Department of Surgery, Vascular Surgery Division (D.T., F.S.), National and Kapodistrian University of Athens Medical School, Greece
| | - Ralf P Brandes
- Centre for Molecular Medicine, Institute for Cardiovascular Physiology (C.R., M.S.L., R.P.B.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Stefanie Dimmeler
- Institute for Cardiovascular Regeneration (L.T., S.D.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy (A.P.), National and Kapodistrian University of Athens Medical School, Greece.,Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece (A.P.)
| | - Stefan Knapp
- Institute for Pharmaceutical Chemistry and Buchmann Institute for Molecular Life Sciences (S.K.), Goethe University, Frankfurt am Main, Germany
| | - Stefan Offermanns
- German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.).,Department of Pharmacology (S.O.), Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Ilka Wittig
- Functional Proteomics (J.Heidler., I.W.), Goethe University, Frankfurt am Main, Germany
| | | | - Fragiska Sigala
- First Propedeutic Department of Surgery, Vascular Surgery Division (D.T., F.S.), National and Kapodistrian University of Athens Medical School, Greece
| | - Ingrid Fleming
- Institute for Vascular Signalling (S-I.B., J.H., J.W., M.K.D., V.R., F.D.L., B.F., S.Z., A.K., A.F.O.J., I.F.), Goethe University, Frankfurt am Main, Germany.,German Center of Cardiovascular Research (DZHK), Partner site RheinMain, Frankfurt am Main, Germany (S-I.B., J.Hu, M.L., C.R., J.W., L.T., V.R., M.S.L., P.G., F.D.L., B.F., S.Z., A.K., A.F.O.J., J.Heidler, R.P.B., S.D., S.O., I.W., I.F.)
| |
Collapse
|
27
|
Piragine E, Calderone V. Pharmacological modulation of the hydrogen sulfide (H 2 S) system by dietary H 2 S-donors: A novel promising strategy in the prevention and treatment of type 2 diabetes mellitus. Phytother Res 2020; 35:1817-1846. [PMID: 33118671 DOI: 10.1002/ptr.6923] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/15/2020] [Accepted: 10/06/2020] [Indexed: 12/25/2022]
Abstract
Type 2 diabetes mellitus (T2DM) represents the most common age-related metabolic disorder, and its management is becoming both a health and economic issue worldwide. Moreover, chronic hyperglycemia represents one of the main risk factors for cardiovascular complications. In the last years, the emerging evidence about the role of the endogenous gasotransmitter hydrogen sulfide (H2 S) in the pathogenesis and progression of T2DM led to increasing interest in the pharmacological modulation of endogenous "H2 S-system". Indeed, H2 S directly contributes to the homeostatic maintenance of blood glucose levels; moreover, it improves impaired angiogenesis and endothelial dysfunction under hyperglycemic conditions. Moreover, H2 S promotes significant antioxidant, anti-inflammatory, and antiapoptotic effects, thus preventing hyperglycemia-induced vascular damage, diabetic nephropathy, and cardiomyopathy. Therefore, H2 S-releasing molecules represent a promising strategy in both clinical management of T2DM and prevention of macro- and micro-vascular complications associated to hyperglycemia. Recently, growing attention has been focused on dietary organosulfur compounds. Among them, garlic polysulfides and isothiocyanates deriving from Brassicaceae have been recognized as H2 S-donors of great pharmacological and nutraceutical interest. Therefore, a better understanding of the therapeutic potential of naturally occurring H2 S-donors may pave the way to a more rational use of these nutraceuticals in the modulation of H2 S homeostasis in T2DM.
Collapse
Affiliation(s)
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Pisa, Italy.,Interdepartmental Research Center Nutrafood "Nutraceuticals and Food for Health", University of Pisa, Pisa, Italy.,Interdepartmental Research Centre of Ageing Biology and Pathology, University of Pisa, Pisa, Italy
| |
Collapse
|
28
|
Paul BD, Snyder SH, Kashfi K. Effects of hydrogen sulfide on mitochondrial function and cellular bioenergetics. Redox Biol 2020; 38:101772. [PMID: 33137711 PMCID: PMC7606857 DOI: 10.1016/j.redox.2020.101772] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2020] [Revised: 10/17/2020] [Accepted: 10/26/2020] [Indexed: 01/06/2023] Open
Abstract
Hydrogen sulfide (H2S) was once considered to have only toxic properties, until it was discovered to be an endogenous signaling molecule. The effects of H2S are dose dependent, with lower concentrations being beneficial and higher concentrations, cytotoxic. This scenario is especially true for the effects of H2S on mitochondrial function, where higher concentrations of the gasotransmitter inhibit the electron transport chain, and lower concentrations stimulate bioenergetics in multiple ways. Here we review the role of H2S in mitochondrial function and its effects on cellular physiology. Hydrogen sulfide (H2S) plays central roles in mitochondrial homeostasis. Both excess H2S and a paucity of H2S have deleterious effects. One of the modes by which H2S signals in mitochondria is by sulfhydrating target proteins. Administering H2S (where scarcity of H2S occurs) or inhibiting H2S production (in case of excess H2S) may be beneficial.
Collapse
Affiliation(s)
- Bindu D Paul
- The Solomon H. Snyder Department of Neuroscience, USA.
| | - Solomon H Snyder
- The Solomon H. Snyder Department of Neuroscience, USA; Department of Psychiatry and Behavioral Sciences, USA; Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, City University of New York School of Medicine, New York, NY, 10031, USA; Graduate Program in Biology, City University of New York Graduate Center, New York, NY, 10016, USA.
| |
Collapse
|
29
|
Sun F, Luo JH, Yue TT, Wang FX, Yang CL, Zhang S, Wang XQ, Wang CY. The role of hydrogen sulphide signalling in macrophage activation. Immunology 2020; 162:3-10. [PMID: 32876334 PMCID: PMC7730026 DOI: 10.1111/imm.13253] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/27/2020] [Accepted: 07/30/2020] [Indexed: 01/05/2023] Open
Abstract
Hydrogen sulphide (H2S) is the latest identified small gaseous mediator enabled by its lipophilic nature to freely permeate the biological membranes. Initially, H2S was recognized by its roles in neuronal activity and vascular relaxation, which makes it an important molecule involved in paracrine signalling pathways. Recently, the immune regulatory function of gasotransmitters, H2S in particular, is increasingly being appreciated. Endogenous H2S level has been linked to macrophage activation, polarization and inflammasome formation. Mechanistically, H2S‐induced protein S‐sulphydration suppresses several inflammatory pathways including NF‐κB and JNK signalling. Moreover, H2S serves as a potent cellular redox regulator to modulate epigenetic alterations and to promote mitochondrial biogenesis in macrophages. Here in this review, we intend to summarize the recent advancements of H2S studies in macrophages, and to discuss with focus on the therapeutic potential of H2S donors by targeting macrophages. The feasibility of H2S signalling component as a macrophage biomarker under disease conditions would be also discussed.
Collapse
Affiliation(s)
- Fei Sun
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Jia-Hui Luo
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Tian-Tian Yue
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Fa-Xi Wang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Chun-Liang Yang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Shu Zhang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Xin-Qiang Wang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Cong-Yi Wang
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China.,Key Laboratory of Pulmonary Diseases of Health Ministry, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China.,Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| |
Collapse
|
30
|
The interplay between oxidative stress and bioenergetic failure in neuropsychiatric illnesses: can we explain it and can we treat it? Mol Biol Rep 2020; 47:5587-5620. [PMID: 32564227 DOI: 10.1007/s11033-020-05590-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2020] [Accepted: 06/12/2020] [Indexed: 12/12/2022]
Abstract
Nitro-oxidative stress and lowered antioxidant defences play a key role in neuropsychiatric disorders such as major depression, bipolar disorder and schizophrenia. The first part of this paper details mitochondrial antioxidant mechanisms and their importance in reactive oxygen species (ROS) detoxification, including details of NO networks, the roles of H2O2 and the thioredoxin/peroxiredoxin system, and the relationship between mitochondrial respiration and NADPH production. The second part highlights and identifies the causes of the multiple pathological sequelae arising from self-amplifying increases in mitochondrial ROS production and bioenergetic failure. Particular attention is paid to NAD+ depletion as a core cause of pathology; detrimental effects of raised ROS and reactive nitrogen species on ATP and NADPH generation; detrimental effects of oxidative and nitrosative stress on the glutathione and thioredoxin systems; and the NAD+-induced signalling cascade, including the roles of SIRT1, SIRT3, PGC-1α, the FOXO family of transcription factors, Nrf1 and Nrf2. The third part discusses proposed therapeutic interventions aimed at mitigating such pathology, including the use of the NAD+ precursors nicotinamide mononucleotide and nicotinamide riboside, both of which rapidly elevate levels of NAD+ in the brain and periphery following oral administration; coenzyme Q10 which, when given with the aim of improving mitochondrial function and reducing nitro-oxidative stress in the brain, may be administered via the use of mitoquinone, which is in essence ubiquinone with an attached triphenylphosphonium cation; and N-acetylcysteine, which is associated with improved mitochondrial function in the brain and produces significant decreases in oxidative and nitrosative stress in a dose-dependent manner.
Collapse
|
31
|
Sun HJ, Wu ZY, Nie XW, Wang XY, Bian JS. Implications of hydrogen sulfide in liver pathophysiology: Mechanistic insights and therapeutic potential. J Adv Res 2020; 27:127-135. [PMID: 33318872 PMCID: PMC7728580 DOI: 10.1016/j.jare.2020.05.010] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Revised: 05/06/2020] [Accepted: 05/07/2020] [Indexed: 02/07/2023] Open
Abstract
Background Over the last several decades, hydrogen sulfide (H2S) has been found to exert multiple physiological functions in mammal systems. The endogenous production of H2S is primarily mediated by cystathione β-synthase (CBS), cystathione γ-lyase (CSE), and 3-mercaptopyruvate sulfurtransferase (3-MST). These enzymes are widely expressed in the liver tissues and regulate hepatic functions by acting on various molecular targets. Aim of Review In the present review, we will highlight the recent advancements in the cellular events triggered by H2S under liver diseases. The therapeutic effects of H2S donors on hepatic diseases will also be discussed. Key Scientific Concepts of Review As a critical regulator of liver functions, H2S is critically involved in the etiology of various liver disorders, such as nonalcoholic steatohepatitis (NASH), hepatic fibrosis, hepatic ischemia/reperfusion (IR) injury, and liver cancer. Targeting H2S-producing enzymes may be a promising strategy for managing hepatic disorders.
Collapse
Key Words
- 3-MP, 3-mercaptopyruvate
- 3-MST, 3-mercaptopyruvate sulfurtransferase
- AGTR1, angiotensin II type 1 receptor
- AMPK, AMP-activated protein kinase
- Akt, protein kinase B
- CAT, cysteine aminotransferase
- CBS, cystathione β-synthase
- CO, carbon monoxide
- COX-2, cyclooxygenase-2
- CSE, cystathione γ-lyase
- CX3CR1, chemokine CX3C motif receptor 1
- Cancer
- DAO, D-amino acid oxidase
- DATS, Diallyl trisulfide
- EGFR, epidermal growth factor receptor
- ERK, extracellular regulated protein kinases
- FAS, fatty acid synthase
- Fibrosis
- H2S, hydrogen sulfide
- HFD, high fat diet
- HO-1, heme oxygenase 1
- Hydrogen sulfide
- IR, ischemia/reperfusion
- Liver disease
- MMP-2, matrix metalloproteinase 2
- NADH, nicotinamide adenine dinucleotide
- NADPH, nicotinamide adenine dinucleotide phosphate
- NAFLD, non-alcoholic fatty liver diseases
- NASH, nonalcoholic steatohepatitis
- NF-κB, nuclear factor-kappa B
- NaHS, sodium hydrosulfide
- Nrf2, nuclear factor erythroid2-related factor 2
- PI3K, phosphatidylinositol 3-kinase
- PLP, pyridoxal 5′-phosphate
- PPG, propargylglycine
- PTEN, phosphatase and tensin homolog deleted on chromosome ten
- SAC, S-allyl-cysteine
- SPRC, S-propargyl-cysteine
- STAT3, signal transducer and activator of transcription 3
- Steatosis
- VLDL, very low density lipoprotein
- mTOR, mammalian target of rapamycin
Collapse
Affiliation(s)
- Hai-Jian Sun
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Zhi-Yuan Wu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Xiao-Wei Nie
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore
| | - Xin-Yu Wang
- Department of Endocrinology, The First Affiliated Hospital of Shenzhen University (Shenzhen Second People's Hospital), Shenzhen 518037, China
| | - Jin-Song Bian
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 117597, Singapore.,National University of Singapore Research Institute, Suzhou 215000, China
| |
Collapse
|
32
|
Popov LD. Mitochondrial biogenesis: An update. J Cell Mol Med 2020; 24:4892-4899. [PMID: 32279443 PMCID: PMC7205802 DOI: 10.1111/jcmm.15194] [Citation(s) in RCA: 438] [Impact Index Per Article: 87.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 03/07/2020] [Accepted: 03/10/2020] [Indexed: 02/06/2023] Open
Abstract
In response to the energy demand triggered by developmental signals and environmental stressors, the cells launch the mitochondrial biogenesis process. This is a self‐renewal route, by which new mitochondria are generated from the ones already existing. Recently, considerable progress has been made in deciphering mitochondrial biogenesis‐related proteins and genes that function in health and in pathology‐related circumstances. However, an outlook on the intracellular mechanisms shared by the main players that drive mitochondrial biogenesis machinery is still missing. Here, we provide such a view by focusing on the following issues: (a) the role of mitochondrial biogenesis in homeostasis of the mitochondrial mass and function, (b) the signalling pathways beyond the induction/promotion, stimulation and inhibition of mitochondrial biogenesis and (c) the therapeutic applications aiming the repair and regeneration of defective mitochondrial biogenesis (in ageing, metabolic diseases, neurodegeneration and cancer). The review is concluded by the perspectives of mitochondrial medicine and research.
Collapse
Affiliation(s)
- Lucia-Doina Popov
- "Nicolae Simionescu" Institute of Cellular Biology and Pathology of the Romanian Academy, Bucharest, Romania
| |
Collapse
|
33
|
Zhang H, Huang Y, Chen S, Tang C, Wang G, Du J, Jin H. Hydrogen sulfide regulates insulin secretion and insulin resistance in diabetes mellitus, a new promising target for diabetes mellitus treatment? A review. J Adv Res 2020; 27:19-30. [PMID: 33318863 PMCID: PMC7728586 DOI: 10.1016/j.jare.2020.02.013] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 02/11/2020] [Accepted: 02/24/2020] [Indexed: 12/28/2022] Open
Abstract
Background Insulin resistance and impaired insulin secretion lead to disorders of glucose metabolism, which contributes to the development of diabetes. Hydrogen sulfide (H2S), a novel gasotransmitter, is found to play important roles in regulation of glucose metabolism homeostasis. Aim of Review This study aimed to summarize and discuss current data about the function of H2S in insulin secretion and insulin resistance regulation as well as the underlying mechanisms. Key Scientific Concepts of Review H2S could be endogenously produced in islet β cells, liver, adipose, skeletal muscles, and the hypothalamus, and regulates local and systemic glucose metabolism. It is reported that H2S suppresses insulin secretion, promotes or reduces the apoptosis of islet β cells. It plays important roles in the regulation of insulin sensitivity in insulin responsive tissues. H2S inhibits glucose uptake and glycogen storage, and promotes or inhibits gluconeogenesis, mitochondrial biogenesis and mitochondrial bioenergetics in the liver. In adipose tissue, several investigators indicated that H2S promoted glucose uptake in adipocytes, while other studies reported that H2S inhibits this process. H2S has also been shown to promote adipogenesis, inhibit lipolysis, and regulate adiponectin and MCP-1 secretion from adipocytes. In skeletal muscle, H2S increases glucose uptake and improves insulin sensitivity. It is also observed that H2S modulates circadian-clock genes in muscle. Hypothalamic CBS/H2S pathway reduces obesity and improves insulin sensitivity via the brain-adipose interaction. Most studies indicated plasma H2S levels decreased in diabetic patients. However, the mechanisms by which H2S regulates systemic glucose metabolism remain unclear. Whether H2S acts as a new promising target for diabetes mellitus treatment merits further studies.
Collapse
Affiliation(s)
- Heng Zhang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China.,Research Unit of Clinical Diagnosis and Treatment of Pediatric Syncope and Cardiovascular Diseases, Chinese Academy of Medical Sciences, China
| | - Selena Chen
- Division of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, United States
| | - Chaoshu Tang
- Department of Physiology and Pathophysiology, Peking University Health Science Centre, Beijing 100091, China.,Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing 100083, China
| | - Guang Wang
- Department of Endocrinology, Beijing Chaoyang Hospital, Capital Medical University, Beijing 100020, China
| | - Junbao Du
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China.,Research Unit of Clinical Diagnosis and Treatment of Pediatric Syncope and Cardiovascular Diseases, Chinese Academy of Medical Sciences, China.,Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing 100083, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First Hospital, Beijing 100034, China.,Research Unit of Clinical Diagnosis and Treatment of Pediatric Syncope and Cardiovascular Diseases, Chinese Academy of Medical Sciences, China.,Key Laboratory of Molecular Cardiology, Ministry of Education, Beijing 100083, China
| |
Collapse
|
34
|
Gheibi S, Samsonov AP, Gheibi S, Vazquez AB, Kashfi K. Regulation of carbohydrate metabolism by nitric oxide and hydrogen sulfide: Implications in diabetes. Biochem Pharmacol 2020; 176:113819. [PMID: 31972170 DOI: 10.1016/j.bcp.2020.113819] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/15/2020] [Indexed: 12/19/2022]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) are two gasotransmitters that are produced in the human body and have a key role in many of the physiological activities of the various organ systems. Decreased NO bioavailability and deficiency of H2S are involved in the pathophysiology of type 2 diabetes and its complications. Restoration of NO levels have favorable metabolic effects in diabetes. The role of H2S in pathophysiology of diabetes is however controversial; H2S production is decreased during development of obesity, diabetes, and its complications, suggesting the potential therapeutic effects of H2S. On the other hand, increased H2S levels disturb the pancreatic β-cell function and decrease insulin secretion. In addition, there appear to be important interactions between NO and H2S at the levels of both biosynthesis and signaling pathways, yet clear an insight into this relationship is lacking. H2S potentiates the effects of NO in the cardiovascular system as well as NO release from its storage pools. Likewise, NO increases the activity and the expression of H2S-generating enzymes. Inhibition of NO production leads to elimination/attenuation of the cardioprotective effects of H2S. Regarding the increasing interest in the therapeutic applications of NO or H2S-releasing molecules in a variety of diseases, particularly in the cardiovascular disorders, much is to be learned about their function in glucose/insulin metabolism, especially in diabetes. The aim of this review is to provide a better understanding of the individual and the interactive roles of NO and H2S in carbohydrate metabolism.
Collapse
Affiliation(s)
- Sevda Gheibi
- Department of Clinical Sciences in Malmö, Unit of Molecular Metabolism, Lund University Diabetes Centre, Clinical Research Center, Malmö University Hospital, Lund University, Malmö, Sweden.
| | - Alan P Samsonov
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Shahsanam Gheibi
- Maternal and Childhood Obesity Research Center, Urmia University of Medical Sciences, Urmia, Iran
| | - Alexandra B Vazquez
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA
| | - Khosrow Kashfi
- Department of Molecular, Cellular and Biomedical Sciences, Sophie Davis School of Biomedical Education, City University of New York School of Medicine, New York, NY, USA; Graduate Program in Biology, City University of New York Graduate Center, New York, NY, USA.
| |
Collapse
|
35
|
Yuan Y, Zhu L, Li L, Liu J, Chen Y, Cheng J, Peng T, Lu Y. S-Sulfhydration of SIRT3 by Hydrogen Sulfide Attenuates Mitochondrial Dysfunction in Cisplatin-Induced Acute Kidney Injury. Antioxid Redox Signal 2019; 31:1302-1319. [PMID: 31218880 DOI: 10.1089/ars.2019.7728] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Aims: Clinical use of cisplatin (Cisp), one of the most widely used, common, and effective chemotherapeutic agents, is limited by its side effects, particularly tubular injury-associated nephrotoxicity. Previous studies suggest that hydrogen sulfide (H2S) alleviates Cisp-induced acute kidney injury (AKI). However, the underlying mechanism remains largely unclear. Results: A single intraperitoneal injection of Cisp is employed to induce AKI, and the mice exhibit severe kidney dysfunction and histological damage at day 4 after Cisp injection. Here, we reported that H2S alleviated Cisp-caused renal toxicity via SIRT3 activation and subsequent improvement of mitochondrial ATP production. Using a biotin-switch assay, we showed that H2S increased S-sulfhydration of SIRT3 and induced deacetylation of its target proteins (OPA1, ATP synthase β, and superoxide dismutase 2). These effects of H2S were associated with a reduction of mitochondrial fragmentation, an increase in ATP generation, and less oxidative injury. Notably, the S-sulfhydration of SIRT3 induced by H2S was abrogated when Cys256, Cys259, Cys280, and Cys283 residues on SIRT3 (two zinc finger domains) were mutated. Innovation and Conclusion: Our data suggest that H2S attenuates Cisp-induced AKI by preventing mitochondrial dysfunction via SIRT3 sulfhydrylation. Antioxid. Redox Signal. 31, 1302-1319.
Collapse
Affiliation(s)
- Yujia Yuan
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu, China.,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lingling Zhu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu, China.,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Lan Li
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu, China.,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingping Liu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu, China.,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China.,Center for Metabolic and Vascular Biology, School for Nutrition and Health Promotion, College of Health Solutions, Arizona State University, Scottsdale, Arizona
| | - Younan Chen
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu, China.,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jingqiu Cheng
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu, China.,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| | - Tianqing Peng
- Critical Illness Research, Lawson Health Research Institute, London Health Sciences Centre, London, Canada.,Departments of Medicine and Pathology, University of Western Ontario, London, Canada
| | - Yanrong Lu
- Key Laboratory of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu, China.,Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
36
|
Murphy B, Bhattacharya R, Mukherjee P. Hydrogen sulfide signaling in mitochondria and disease. FASEB J 2019; 33:13098-13125. [PMID: 31648556 PMCID: PMC6894098 DOI: 10.1096/fj.201901304r] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/17/2019] [Indexed: 02/06/2023]
Abstract
Hydrogen sulfide can signal through 3 distinct mechanisms: 1) reduction and/or direct binding of metalloprotein heme centers, 2) serving as a potent antioxidant through reactive oxygen species/reactive nitrogen species scavenging, or 3) post-translational modification of proteins by addition of a thiol (-SH) group onto reactive cysteine residues: a process known as persulfidation. Below toxic levels, hydrogen sulfide promotes mitochondrial biogenesis and function, thereby conferring protection against cellular stress. For these reasons, increases in hydrogen sulfide and hydrogen sulfide-producing enzymes have been implicated in several human disease states. This review will first summarize our current understanding of hydrogen sulfide production and metabolism, as well as its signaling mechanisms; second, this work will detail the known mechanisms of hydrogen sulfide in the mitochondria and the implications of its mitochondrial-specific impacts in several pathologic conditions.-Murphy, B., Bhattacharya, R., Mukherjee, P. Hydrogen sulfide signaling in mitochondria and disease.
Collapse
Affiliation(s)
- Brennah Murphy
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Priyabrata Mukherjee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
37
|
Sun X, Zhao D, Lu F, Peng S, Yu M, Liu N, Sun Y, Du H, Wang B, Chen J, Dong S, Lu F, Zhang W. Hydrogen sulfide regulates muscle RING finger-1 protein S-sulfhydration at Cys 44 to prevent cardiac structural damage in diabetic cardiomyopathy. Br J Pharmacol 2019; 177:836-856. [PMID: 30734268 DOI: 10.1111/bph.14601] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2018] [Revised: 12/04/2018] [Accepted: 12/19/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Hydrogen sulfide (H2 S) plays important roles as a gasotransmitter in pathologies. Increased expression of the E3 ubiquitin ligase, muscle RING finger-1 (MuRF1), may be involved in diabetic cardiomyopathy. Here we have investigated whether and how exogenous H2 S alleviates cardiac muscle degradation through modifications of MuRF1 S-sulfhydration in db/db mice. EXPERIMENTAL APPROACH Neonatal rat cardiomyocytes were treated with high glucose (40 mM), oleate (100 μM), palmitate (400 μM), and NaHS (100 μM) for 72 hr. MuRF1 was silenced with siRNA technology and mutation at Cys44 . Endoplasmic reticulum stress markers, MuRF1 expression, and ubiquitination level were measured. db/db mice were injected with NaHS (39 μmol·kg-1 ) for 20 weeks. Echocardiography, cardiac ultrastructure, cystathionine-γ-lyase, cardiac structure proteins expression, and S-sulfhydration production were measured. KEY RESULTS H2 S levels and cystathionine-γ-lyase protein expression in myocardium were decreased in db/db mice. Exogenous H2 S reversed endoplasmic reticulum stress, including impairment of the function of cardiomyocytes and structural damage in db/db mice. Exogenous H2 S could suppress the levels of myosin heavy chain 6 and myosin light chain 2 ubiquitination in cardiac tissues of db/db mice, and MuRF1 was modified by S-sulfhydration, following treatment with exogenous H2 S, to reduce the interaction between MuRF1 and myosin heavy chain 6 and myosin light chain 2. CONCLUSIONS AND IMPLICATIONS Our findings suggest that H2 S regulates MuRF1 S-sulfhydration at Cys44 to prevent myocardial degradation in the cardiac tissues of db/db mice. LINKED ARTICLES This article is part of a themed section on Hydrogen Sulfide in Biology & Medicine. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v177.4/issuetoc.
Collapse
Affiliation(s)
- Xiaojiao Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Dechao Zhao
- Department of Cardiology, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Fangping Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shuo Peng
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Miao Yu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Ning Liu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Yu Sun
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Haining Du
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Bingzhu Wang
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Jian Chen
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Shiyun Dong
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Fanghao Lu
- Department of Pathophysiology, Harbin Medical University, Harbin, China
| | - Weihua Zhang
- Department of Pathophysiology, Harbin Medical University, Harbin, China.,Key Laboratory of Cardiovascular Medicine Research, Harbin Medical University Ministry of Education, Harbin, China
| |
Collapse
|
38
|
Sakai K, Katsumi H, Kamano K, Yamauchi K, Hajima A, Morishita M, Sakane T, Yamamoto A. Hepatic and Intrahepatic Targeting of Hydrogen Sulfide Prodrug by Bioconjugation. Biol Pharm Bull 2019; 42:273-279. [PMID: 30713258 DOI: 10.1248/bpb.b18-00773] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Hydrogen sulfide (H2S) is an endogenous gaseous transmitter known to play an important role in biological functions. For the hepatic and intrahepatic targeting of H2S prodrug at the cellular level, we developed two types of sulfo-albumins, in which five sulfide groups (source of H2S) were covalently bound to succinylated (Suc) or galactosylated (Gal) bovine serum albumin (BSA). Sulfo-BSA-Suc and polyethylene glycol (PEG)-Sulfo-BSA-Gal, both released H2S in the 5 mM glutathione solution, but not in the plasma. Sulfo-BSA-Suc and PEG-Sulfo-BSA-Gal were taken up by RAW264.7 cells (mouse macrophage-like cells) and Hep G2 cells (human hepatocellular carcinoma cells), respectively, and H2S was released. These results indicate that Sulfo-BSA-Suc and PEG -Sulfo-BSA-Gal selectively released H2S intracellularly. In a biodistribution study, up to 80% of 111In-labeled Sulfo-BSA-Suc and PEG-Sulfo-BSA-Gal rapidly accumulated in the liver, 30 min after intravenous injection in mice. Furthermore, 111In-labeled Sulfo-BSA-Suc and PEG-Sulfo-BSA-Gal predominantly accumulated in liver nonparenchymal (endothelial cells and Kupffer cells) and parenchymal cells (hepatocytes), respectively. These findings suggest that targeted delivery of H2S prodrug to a specific type of liver cells was successfully achieved by bioconjugation.
Collapse
Affiliation(s)
- Kosuke Sakai
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| | | | - Kentaro Kamano
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| | - Kiyo Yamauchi
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| | - Ayuko Hajima
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| | | | - Toshiyasu Sakane
- Department of Biopharmaceutics, Kyoto Pharmaceutical University.,Department of Pharmaceutical Technology, Kobe Pharmaceutical University
| | - Akira Yamamoto
- Department of Biopharmaceutics, Kyoto Pharmaceutical University
| |
Collapse
|
39
|
Hydrogen Sulfide as a Novel Regulatory Factor in Liver Health and Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:3831713. [PMID: 30805080 PMCID: PMC6360590 DOI: 10.1155/2019/3831713] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 11/29/2018] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S), a colorless gas smelling of rotten egg, has long been recognized as a toxic gas and environment pollutant. However, increasing evidence suggests that H2S acts as a novel gasotransmitter and plays important roles in a variety of physiological and pathological processes in mammals. H2S is involved in many hepatic functions, including the regulation of oxidative stress, glucose and lipid metabolism, vasculature, mitochondrial function, differentiation, and circadian rhythm. In addition, H2S contributes to the pathogenesis and treatment of a number of liver diseases, such as hepatic fibrosis, liver cirrhosis, liver cancer, hepatic ischemia/reperfusion injury, nonalcoholic fatty liver disease/nonalcoholic steatohepatitis, hepatotoxicity, and acute liver failure. In this review, the biosynthesis and metabolism of H2S in the liver are summarized and the role and mechanism of H2S in liver health and disease are further discussed.
Collapse
|
40
|
Theaflavins Improve Insulin Sensitivity through Regulating Mitochondrial Biosynthesis in Palmitic Acid-Induced HepG2 Cells. Molecules 2018; 23:molecules23123382. [PMID: 30572687 PMCID: PMC6320999 DOI: 10.3390/molecules23123382] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 12/16/2018] [Accepted: 12/17/2018] [Indexed: 12/31/2022] Open
Abstract
Theaflavins, the characteristic and bioactive polyphenols in black tea, possess the potential improving effects on insulin resistance-associated metabolic abnormalities, including obesity and type 2 diabetes mellitus. However, the related molecular mechanisms are still unclear. In this research, we investigated the protective effects of theaflavins against insulin resistance in HepG2 cells induced by palmitic acid. Theaflavins significantly increased glucose uptake of insulin-resistant cells at noncytotoxic doses. This activity was mediated by upregulating the total and membrane bound glucose transporter 4 protein expressions, increasing the phosphor-Akt (Ser473) level, and decreasing the phosphorylation of IRS-1 at Ser307. Moreover, theaflavins were found to enhance the mitochondrial DNA copy number, down-regulate the PGC-1β mRNA level and increase the PRC mRNA expression. Mdivi-1, a selective mitochondrial division inhibitor, could attenuate TFs-induced promotion of glucose uptake in insulin-resistant HepG2 cells. Taken together, these results suggested that theaflavins could improve hepatocellular insulin resistance induced by free fatty acids, at least partly through promoting mitochondrial biogenesis. Theaflavins are promising functional food ingredients and medicines for improving insulin resistance-related disorders.
Collapse
|
41
|
Abstract
SIGNIFICANCE Among many endogenous mediators, the gasotransmitter hydrogen sulfide (H2S) plays an important role in the regulation of glucose homeostasis. In this article we discuss different functional roles of H2S in several metabolic organs/tissues required in the maintenance of glucose homeostasis. Recent Advances: New evidence has emerged revealing the insulin sensitizing role of H2S in adipose tissue and skeletal muscle biology. In addition, H2S was demonstrated to be a potent stimulator of gluconeogenesis via the induction and stimulation of various glucose-producing pathways in the liver. CRITICAL ISSUES Similar to its other physiological effects, H2S exhibits paradoxical characteristics in the regulation of glucose homeostasis: (1) H2S stimulates glucose production via activation of gluconeogenesis and glycogenolysis in hepatocytes, yet inhibits lipolysis in adipocytes; (2) H2S stimulates glucose uptake into adipocytes and skeletal muscle but inhibits glucose uptake into hepatocytes; (3) H2S inhibits insulin secretion from pancreatic β cells, yet sensitizes insulin signaling and insulin-triggered response in adipose tissues and skeletal muscle. It is also unclear the impact H2S may have on glucose metabolism and utilization by other vital organs, such as the brain. FUTURE DIRECTIONS Recent reports and ongoing studies lay the foundation for a general, although highly incomplete, understanding of the effect of H2S on regulating glucose homeostasis. In this review, we describe the molecular mechanisms and physiological outcomes of the gasotransmitter H2S on organs and tissues required for homeostatic maintenance of blood glucose. Future directions highlighting the H2S-mediated homeostatic control of glucose metabolism under physiological and insulin-resistant conditions are also discussed. Antioxid. Redox Signal. 28, 1463-1482.
Collapse
Affiliation(s)
- Ashley Untereiner
- 1 Department of Anesthesiology, University of Texas Medical Branch , Galveston, Texas
| | - Lingyun Wu
- 2 Cardiovascular & Metabolic Research Unit and School of Human Kinetics, Laurentian University , Sudbury, Canada .,3 Health Sciences North Research Institute , Sudbury, Canada
| |
Collapse
|
42
|
Hydrogen sulfide in the regulation of insulin secretion and insulin sensitivity: Implications for the pathogenesis and treatment of diabetes mellitus. Biochem Pharmacol 2018; 149:60-76. [DOI: 10.1016/j.bcp.2018.01.004] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 01/02/2018] [Indexed: 01/04/2023]
|
43
|
Shimizu Y, Polavarapu R, Eskla KL, Nicholson CK, Koczor CA, Wang R, Lewis W, Shiva S, Lefer DJ, Calvert JW. Hydrogen sulfide regulates cardiac mitochondrial biogenesis via the activation of AMPK. J Mol Cell Cardiol 2018; 116:29-40. [PMID: 29408195 DOI: 10.1016/j.yjmcc.2018.01.011] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 01/03/2018] [Accepted: 01/18/2018] [Indexed: 12/13/2022]
Abstract
BACKGROUND Hydrogen sulfide (H2S) is an important regulator of mitochondrial bioenergetics, but its role in regulating mitochondrial biogenesis is not well understood. Using both genetic and pharmacological approaches, we sought to determine if H2S levels directly influenced cardiac mitochondrial content. RESULTS Mice deficient in the H2S-producing enzyme, cystathionine γ-lyase (CSE KO) displayed diminished cardiac mitochondrial content when compared to wild-type hearts. In contrast, mice overexpressing CSE (CSE Tg) and mice supplemented with the orally active H2S-releasing prodrug, SG-1002, displayed enhanced cardiac mitochondrial content. Additional analysis revealed that cardiac H2S levels influenced the nuclear localization and transcriptional activity of peroxisome proliferator-activated receptor γ coactivator 1α (PGC1α) with higher levels having a positive influence and lower levels having a negative influence. Studies aimed at evaluating the underlying mechanisms found that H2S required AMP-activated protein kinase (AMPK) to induce PGC1α signaling and mitochondrial biogenesis. Finally, we found that restoring H2S levels with SG-1002 in the setting of heart failure increased cardiac mitochondrial content, improved mitochondrial respiration, improved ATP production efficiency, and improved cardiac function. CONCLUSIONS Together, these results suggest that hydrogen sulfide is an important regulator of cardiac mitochondrial content and establishes that exogenous hydrogen sulfide can induce mitochondrial biogenesis via an AMPK-PGC1α signaling cascade.
Collapse
Affiliation(s)
- Yuuki Shimizu
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Rohini Polavarapu
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Kattri-Liis Eskla
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA, USA
| | - Chad K Nicholson
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Rui Wang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Sudbury, Canada
| | - William Lewis
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Sruti Shiva
- Department of Pharmacology & Chemical Biology, Vascular Medicine Institute, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - David J Lefer
- Cardiovascular Center of Excellence, Department of Pharmacology, LSU Health Sciences Center, New Orleans, LA, USA
| | - John W Calvert
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, Atlanta, GA, USA.
| |
Collapse
|
44
|
Lee SR, Nilius B, Han J. Gaseous Signaling Molecules in Cardiovascular Function: From Mechanisms to Clinical Translation. Rev Physiol Biochem Pharmacol 2018; 174:81-156. [PMID: 29372329 DOI: 10.1007/112_2017_7] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Carbon monoxide (CO), hydrogen sulfide (H2S), and nitric oxide (NO) constitute endogenous gaseous molecules produced by specific enzymes. These gases are chemically simple, but exert multiple effects and act through shared molecular targets to control both physiology and pathophysiology in the cardiovascular system (CVS). The gases act via direct and/or indirect interactions with each other in proteins such as heme-containing enzymes, the mitochondrial respiratory complex, and ion channels, among others. Studies of the major impacts of CO, H2S, and NO on the CVS have revealed their involvement in controlling blood pressure and in reducing cardiac reperfusion injuries, although their functional roles are not limited to these conditions. In this review, the basic aspects of CO, H2S, and NO, including their production and effects on enzymes, mitochondrial respiration and biogenesis, and ion channels are briefly addressed to provide insight into their biology with respect to the CVS. Finally, potential therapeutic applications of CO, H2S, and NO with the CVS are addressed, based on the use of exogenous donors and different types of delivery systems.
Collapse
Affiliation(s)
- Sung Ryul Lee
- Department of Convergence Biomedical Science, Cardiovascular and Metabolic Disease Center, College of Medicine, Inje University, Busan, Republic of Korea
| | - Bernd Nilius
- Laboratory of Ion Channel Research, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium
| | - Jin Han
- National Research Laboratory for Mitochondrial Signaling, Department of Physiology, Department of Health Sciences and Technology, BK21 Plus Project Team, Cardiovascular and Metabolic Disease Center, Inje University, Busan, Republic of Korea.
| |
Collapse
|
45
|
Shuang T, Fu M, Yang G, Wu L, Wang R. The interaction of IGF-1/IGF-1R and hydrogen sulfide on the proliferation of mouse primary vascular smooth muscle cells. Biochem Pharmacol 2017; 149:143-152. [PMID: 29248598 DOI: 10.1016/j.bcp.2017.12.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2017] [Accepted: 12/12/2017] [Indexed: 11/25/2022]
Abstract
Hydrogen sulfide (H2S) is mostly produced by cystathionine-gamma-lyase (CSE) in vascular system and it inhibits the proliferation of vascular smooth muscle cells (SMCs). Insulin-like growth factor-1 (IGF-1), via its receptor (IGF-1R), exerts multiple physiological and pathophysiological effects on the vasculature, including stimulating SMC proliferation and migration, and inhibiting SMC apoptosis. Since H2S and IGF-1/IGF-1R have opposite effects on SMC proliferation, it becomes imperative to better understand the interaction of these two signaling mechanisms on SMC proliferation. SMCs isolated from small mesenteric arteries of CSE knockout (KO) and wild-type (WT) mice were used in the present study. The effects of IGF-1 and H2S on SMC proliferation were evaluated with 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and bromodeoxyuridine (BrdU) assays. Protein expression was determined by western blot, and H2S-induced protein S-sulfhydration was assessed with a modified biotin switch assay. We found that IGF-1 dose-dependently increased the proliferation of both WT-SMCs and KO-SMCs, and this effect was more significant in KO-SMCs. Supplement of sodium hydrosulfide (NaHS) inhibited IGF-1-induced cell proliferation, while this effect was abolished by blocking IGF-1/IGF-1R signaling with picropodophyllin (PPP) or knocking out of the expression of IGF-1R. H2S significantly down-regulates the expression of IGF-1R, stimulates IGF-1R S-sulfhydration, and attenuates the binding of IGF-1 with IGF-1R. This study provides novel insight on the involvement of IGF-1/IGF-1R in H2S-inhibited SMC proliferation and suggests H2S-based innovative treatment strategies for proliferative cardiovascular diseases such as atherosclerosis.
Collapse
Affiliation(s)
- Tian Shuang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Canada; School of Human Kinetics, Laurentian University, Canada; Department of Biology, Laurentian University, Canada; Health Sciences North Research Institute, Sudbury, Ontario, Canada
| | - Ming Fu
- Cardiovascular and Metabolic Research Unit, Laurentian University, Canada; School of Human Kinetics, Laurentian University, Canada; Health Sciences North Research Institute, Sudbury, Ontario, Canada
| | - Guangdong Yang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Canada; Department of Chemistry and Biochemistry, Laurentian University, Ontario, Canada
| | - Lingyun Wu
- Cardiovascular and Metabolic Research Unit, Laurentian University, Canada; School of Human Kinetics, Laurentian University, Canada; Health Sciences North Research Institute, Sudbury, Ontario, Canada
| | - Rui Wang
- Cardiovascular and Metabolic Research Unit, Laurentian University, Canada; Department of Biology, Laurentian University, Canada.
| |
Collapse
|
46
|
Untereiner AA, Pavlidou A, Druzhyna N, Papapetropoulos A, Hellmich MR, Szabo C. Drug resistance induces the upregulation of H 2S-producing enzymes in HCT116 colon cancer cells. Biochem Pharmacol 2017; 149:174-185. [PMID: 29061341 DOI: 10.1016/j.bcp.2017.10.007] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Accepted: 10/18/2017] [Indexed: 01/01/2023]
Abstract
Hydrogen sulfide (H2S) production in colon cancer cells supports cellular bioenergetics and proliferation. The aim of the present study was to investigate the alterations in H2S homeostasis during the development of resistance to 5-fluorouracil (5-FU), a commonly used chemotherapeutic agent. A 5-FU-resistant HCT116 human colon cancer cell line was established by serial passage in the presence of increasing 5-FU concentrations. The 5-FU-resistant cells also demonstrated a partial resistance to an unrelated chemotherapeutic agent, oxaliplatin. Compared to parental cells, the 5-FU-resistant cells rely more on oxidative phosphorylation than glycolysis for bioenergetic function. There was a significant increase in the expression of the drug-metabolizing cytochrome P450 enzymes CYP1A2 and CYP2A6 in 5-FU-resistant cells. The CYP450 inhibitor phenylpyrrole enhanced 5-FU-induced cytotoxicity in 5-FU-resistant cells. Two major H2S-generating enzymes, cystathionine-β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST) were upregulated in the 5-FU-resistant cells. 5-FU-resistant cells exhibited decreased sensitivity to the CBS inhibitor aminooxyacetate (AOAA) in terms of suppression of cell viability, inhibition of cell proliferation and inhibition of oxidative phosphorylation. However, 5FU-resistant cells remained sensitive to the antiproliferative effect of benserazide (a recently identified, potentially repurposable CBS inhibitor). Taken together, the current data suggest that 5-FU resistance in HCT116 cells is associated with the upregulation of drug-metabolizing enzymes and an enhancement of endogenous H2S production. The anticancer effect of prototypical H2S biosynthesis inhibitor AOAA is impaired in 5-FU-resistant cells, but benserazide remains efficacious. Pharmacological approaches aimed at restoring the sensitivity of 5-FU-resistant cells to chemotherapeutic agents may be useful in the formulation of novel therapeutic strategies against colorectal cancer.
Collapse
Affiliation(s)
- Ashley A Untereiner
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Athanasia Pavlidou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece
| | - Nadiya Druzhyna
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Greece; Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| | - Mark R Hellmich
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| |
Collapse
|
47
|
Zhang D, Du J, Tang C, Huang Y, Jin H. H 2S-Induced Sulfhydration: Biological Function and Detection Methodology. Front Pharmacol 2017; 8:608. [PMID: 28932194 PMCID: PMC5592224 DOI: 10.3389/fphar.2017.00608] [Citation(s) in RCA: 121] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2017] [Accepted: 08/22/2017] [Indexed: 12/11/2022] Open
Abstract
At appropriate concentrations, hydrogen sulfide, a well-known gasotransmitter, plays important roles in both physiology and pathophysiology. Increasing evidence suggests that modifying thiol groups of specific cysteines in target proteins via sulfhydration or persulfidation is one of the important mechanisms responsible for the biological functions of hydrogen sulfide. A variety of key proteins of different cellular pathways in mammals have been reported to be sulfhydrated by hydrogen sulfide to participate and regulate the processes of cell survival/death, cell differentiation, cell proliferation/hypertrophy, cellular metabolism, mitochondrial bioenergetics/biogenesis, endoplasmic reticulum stress, vasorelaxtion, inflammation, oxidative stress, etc. Moreover, S-sulfhydration also exerts many biological functions through the cross-talk with other post-translational modifications including phosphorylation, S-nitrosylation and tyrosine nitration. This review summarizes recent studies of hydrogen sulfide-induced sulfhydration as a posttranslational modification, an important biological function of hydrogen sulfide, and sulfhydrated proteins are introduced. Additionally, we discuss the main methods of detecting sulfhydration of proteins.
Collapse
Affiliation(s)
- Da Zhang
- Department of Pediatrics, Peking University First HospitalBeijing, China
| | - Junbao Du
- Department of Pediatrics, Peking University First HospitalBeijing, China.,Key Laboratory of Molecular Cardiology, Ministry of EducationBeijing, China
| | - Chaoshu Tang
- Key Laboratory of Molecular Cardiology, Ministry of EducationBeijing, China.,Department of Physiology and Pathophysiology, Peking University Health Science CenterBeijing, China
| | - Yaqian Huang
- Department of Pediatrics, Peking University First HospitalBeijing, China
| | - Hongfang Jin
- Department of Pediatrics, Peking University First HospitalBeijing, China
| |
Collapse
|