1
|
Matsui M, Lynch LE, Distefano I, Galante A, Gade AR, Wang HG, Gómez-Banoy N, Towers P, Sinden DS, Wei EQ, Barnett AS, Johnson K, Lima R, Rubio-Navarro A, Li AK, Marx SO, McGraw TE, Thornton PS, Timothy KW, Lo JC, Pitt GS. Multiple beta cell-independent mechanisms drive hypoglycemia in Timothy syndrome. Nat Commun 2024; 15:8980. [PMID: 39420001 PMCID: PMC11487186 DOI: 10.1038/s41467-024-52885-3] [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: 11/22/2022] [Accepted: 09/25/2024] [Indexed: 10/19/2024] Open
Abstract
The canonical G406R mutation that increases Ca2+ influx through the CACNA1C-encoded CaV1.2 Ca2+ channel underlies the multisystem disorder Timothy syndrome (TS), characterized by life-threatening arrhythmias. Severe episodic hypoglycemia is among the poorly characterized non-cardiac TS pathologies. While hypothesized from increased Ca2+ influx in pancreatic beta cells and consequent hyperinsulinism, this hypoglycemia mechanism is undemonstrated because of limited clinical data and lack of animal models. We generated a CaV1.2 G406R knockin mouse model that recapitulates key TS features, including hypoglycemia. Unexpectedly, these mice do not show hyperactive beta cells or hyperinsulinism in the setting of normal intrinsic beta cell function, suggesting dysregulated glucose homeostasis. Patient data confirm the absence of hyperinsulinism. We discover multiple alternative contributors, including perturbed counterregulatory hormone responses with defects in glucagon secretion and abnormal hypothalamic control of glucose homeostasis. These data provide new insights into contributions of CaV1.2 channels and reveal integrated consequences of the mutant channels driving life-threatening events in TS.
Collapse
Affiliation(s)
- Maiko Matsui
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Lauren E Lynch
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Isabella Distefano
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Allison Galante
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Aravind R Gade
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Hong-Gang Wang
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Nicolas Gómez-Banoy
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Patrick Towers
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Daniel S Sinden
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Eric Q Wei
- Department of Medicine, MSRB II, 2 Genome Ct, Duke University Medical Center, Durham, NC, 27710, USA
| | - Adam S Barnett
- Department of Medicine, MSRB II, 2 Genome Ct, Duke University Medical Center, Durham, NC, 27710, USA
| | - Kenneth Johnson
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Renan Lima
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Alfonso Rubio-Navarro
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Ang K Li
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
| | - Steven O Marx
- Division of Cardiology, Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, 622 W 168th St, PH-3 Center, New York, NY, USA
- Department of Pharmacology, Vagelos College of Physicians and Surgeons, Columbia University, 622 W 168th St, PH-3 Center, New York, NY, USA
| | - Timothy E McGraw
- Department of Biochemistry, Weill Cornell Medical College, 1300 York Ave, New York, NY, 10065, USA
| | - Paul S Thornton
- Division of Endocrinology and Diabetes, Cook Children's Medical Center, 801 7th Ave, Fort Worth, TX, 76104, USA
| | - Katherine W Timothy
- Children's Hospital Boston, Harvard Medical School, 300 Longwood Ave., Boston, MA, 02115, USA
| | - James C Lo
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Weill Center for Metabolic Health, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA
- Division of Cardiology, Department of Medicine, Weill Cornell Medicine, 413 E. 69th St, New York, NY, 10021, USA
| | - Geoffrey S Pitt
- Cardiovascular Research Institute, Weill Cornell Medicine, 413 E. 69th St., New York, NY, 10021, USA.
| |
Collapse
|
2
|
Mahapatra C, Thakkar R, Kumar R. Modulatory Impact of Oxidative Stress on Action Potentials in Pathophysiological States: A Comprehensive Review. Antioxidants (Basel) 2024; 13:1172. [PMID: 39456426 PMCID: PMC11504047 DOI: 10.3390/antiox13101172] [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: 08/06/2024] [Revised: 09/12/2024] [Accepted: 09/20/2024] [Indexed: 10/28/2024] Open
Abstract
Oxidative stress, characterized by an imbalance between the production of reactive oxygen species (ROS) and the body's antioxidant defenses, significantly affects cellular function and viability. It plays a pivotal role in modulating membrane potentials, particularly action potentials (APs), essential for properly functioning excitable cells such as neurons, smooth muscles, pancreatic beta cells, and myocytes. The interaction between oxidative stress and AP dynamics is crucial for understanding the pathophysiology of various conditions, including neurodegenerative diseases, cardiac arrhythmias, and ischemia-reperfusion injuries. This review explores how oxidative stress influences APs, focusing on alterations in ion channel biophysics, gap junction, calcium dynamics, mitochondria, and Interstitial Cells of Cajal functions. By integrating current research, we aim to elucidate how oxidative stress contributes to disease progression and discuss potential therapeutic interventions targeting this interaction.
Collapse
Affiliation(s)
- Chitaranjan Mahapatra
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA 94158, USA
| | - Ravindra Thakkar
- California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720, USA
| | - Ravinder Kumar
- Department of Pathology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
3
|
Quan X, Zhang M, Qiao Z, Kou X, Xue Q, Wang J, Li L. Nitric oxide and ion channels mediate L-cysteine-induced inhibition of colonic smooth muscle contraction. J Muscle Res Cell Motil 2024; 45:11-20. [PMID: 38141146 DOI: 10.1007/s10974-023-09664-2] [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: 08/13/2023] [Accepted: 12/07/2023] [Indexed: 12/24/2023]
Abstract
Previous studies have suggested that L-cysteine regulates gut motility through hydrogen sulfide. However, the mechanisms involved in the L-cysteine-induced response have not been extensively studied. This study aimed to investigate the underlying mechanisms of action of L-cysteine on spontaneous contraction of rat colon. Longitudinal and circular muscle strips from rat middle colon were prepared to measure the spontaneous contractile activities of colon in an organ bath system. Whole-cell voltage-clamp techniques were applied to record the currents of L-type voltage-dependent Ca2+ channels (VDCCs) and voltage-gated K+ channels (Kv) in isolated smooth muscle cells (SMCs) from colon. L-cysteine inhibited the spontaneous contraction of longitudinal and circular muscle strips from the rat colon in a concentration-dependent manner. The inhibition induced by L-cysteine was significantly decreased by inhibitors of H2S synthesis (p < 0.05). Furthermore, the suppression induced by L-cysteine was partially attenuated by tetrodotoxin, L-NNA and glibenclamide (p < 0.05). Whole-cell voltage-clamp recordings showed that L-cysteine caused a remarkable reduction in the peak currents of VDCCs and significantly increased the membrane currents of Kv channels in isolated SMCs (p < 0.05). We concluded that L-cysteine inhibits the contractile activities of smooth muscle strips from the rat colon. The relaxation in response to L-cysteine may be in part mediated by a nitrergic pathway and by inhibiting the VDCCs in combination with a direct activation of the KV channels and KATP channels.
Collapse
Affiliation(s)
- Xiaojing Quan
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | - Min Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | - Zhaojun Qiao
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | - Xuan Kou
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | - Qiong Xue
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | - Jinhai Wang
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China
| | - Lu Li
- Department of Gastroenterology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710004, Shaanxi Province, China.
| |
Collapse
|
4
|
Zou J, Yuan Z, Chen X, Chen Y, Yao M, Chen Y, Li X, Chen Y, Ding W, Xia C, Zhao Y, Gao F. Hydrogen sulfide responsive nanoplatforms: Novel gas responsive drug delivery carriers for biomedical applications. Asian J Pharm Sci 2024; 19:100858. [PMID: 38362469 PMCID: PMC10867614 DOI: 10.1016/j.ajps.2023.100858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 07/30/2023] [Accepted: 10/06/2023] [Indexed: 02/17/2024] Open
Abstract
Hydrogen sulfide (H2S) is a toxic, essential gas used in various biological and physical processes and has been the subject of many targeted studies on its role as a new gas transmitter. These studies have mainly focused on the production and pharmacological side effects caused by H2S. Therefore, effective strategies to remove H2S has become a key research topic. Furthermore, the development of novel nanoplatforms has provided new tools for the targeted removal of H2S. This paper was performed to review the association between H2S and disease, related H2S inhibitory drugs, as well as H2S responsive nanoplatforms (HRNs). This review first analyzed the role of H2S in multiple tissues and conditions. Second, common drugs used to eliminate H2S, as well as their potential for combination with anticancer agents, were summarized. Not only the existing studies on HRNs, but also the inhibition H2S combined with different therapeutic methods were both sorted out in this review. Furthermore, this review provided in-depth analysis of the potential of HRNs about treatment or detection in detail. Finally, potential challenges of HRNs were proposed. This study demonstrates the excellent potential of HRNs for biomedical applications.
Collapse
Affiliation(s)
- Jiafeng Zou
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Zeting Yuan
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiaojie Chen
- Key Laboratory of Neuropharmacology and Translational Medicine of Zhejiang Province, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - You Chen
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Min Yao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yang Chen
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Xiang Li
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yi Chen
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Wenxing Ding
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Chuanhe Xia
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Yuzheng Zhao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
- CAS Center for Excellence in Brain Science, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing 100050, China
| | - Feng Gao
- Shanghai Frontier Science Research Base of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of Functional Materials Chemistry, East China University of Science and Technology, Shanghai 200237, China
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai 200237, China
| |
Collapse
|
5
|
Mustafa AM, Shaheen AM, Zaki HF, Rabie MA. Nicorandil and carvedilol mitigates motor deficits in experimental autoimmune encephalomyelitis-induced multiple sclerosis: Role of TLR4/TRAF6/MAPK/NF-κB signalling cascade. Int Immunopharmacol 2024; 127:111387. [PMID: 38134593 DOI: 10.1016/j.intimp.2023.111387] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 11/27/2023] [Accepted: 12/12/2023] [Indexed: 12/24/2023]
Abstract
Multiple sclerosis (MS) is an inflammatory demyelinating neurodegenerative disease that negatively affects neurotransmission. It can be pathologically mimicked by experimental autoimmune encephalomyelitis (EAE) animal model. ATP-sensitive potassium channels (KATP) plays a crucial role in the control of neuronal damage, however their role in MS are still obscure. Additionally, Carvedilol showed a promising neuroprotective activity against several neurological disorders. Therefore, the present study aimed to investigate the potential neuroprotective effect of KATP channel opener (nicorandil) as well as α and β adrenoceptor antagonist (Carvedilol) against EAE induced neurodegeneration in mice. Mice was treated with nicorandil (6 mg/kg/day; p.o.) and carvedilol (10 mg/kg/day; p.o.) for 14 days. Nicorandil and carvedilol showed improvement in clinical scoring, behaviour and motor coordination as established by histopathological investigation and immunohistochemical detection of MBP. Furthermore, both treatments downregulated the protein expression of TLR4/ MYD88/TRAF6 signalling cascade with downstream inhibition of (pT183/Y185)-JNK/p38 (pT180/Y182)-MAPK axis leading to reduction of neuroinflammatory status, as witnessed by reduction of NF-κB, TNF-α, IL-1β and IL-6 contents. Moreover, nicorandil and carvedilol attenuated oxidative damage by increasing Nrf2 content and SOD activity together with reduction of MDA content. In addition, an immunomodulating effect via inhibiting the gene expression of CD4, TGF-β, and IL-17 as well as TGF-β, IL-17, and IL-23 contents along with anti-apoptotic effect by decreasing Bax protein expression and Caspase-3 content and increasing Bcl-2 protein expression was observed with nicorandil and carvedilol treatments. In conclusion, nicorandil and carvedilol exerted a neuroprotective activity against EAE induced neuronal loss via inhibition of TLR4/MYD88/TRAF6/JNK/p38-MAPK axis besides antioxidant and anti-apoptotic effects.
Collapse
Affiliation(s)
- Aya M Mustafa
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Aya M Shaheen
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Egyptian Russian University, Cairo, Egypt
| | - Hala F Zaki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Mostafa A Rabie
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| |
Collapse
|
6
|
Łoboda A, Dulak J. Cardioprotective Effects of Hydrogen Sulfide and Its Potential Therapeutic Implications in the Amelioration of Duchenne Muscular Dystrophy Cardiomyopathy. Cells 2024; 13:158. [PMID: 38247849 PMCID: PMC10814317 DOI: 10.3390/cells13020158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 01/23/2024] Open
Abstract
Hydrogen sulfide (H2S) belongs to the family of gasotransmitters and can modulate a myriad of biological signaling pathways. Among others, its cardioprotective effects, through antioxidant, anti-inflammatory, anti-fibrotic, and proangiogenic activities, are well-documented in experimental studies. Cardiorespiratory failure, predominantly cardiomyopathy, is a life-threatening complication that is the number one cause of death in patients with Duchenne muscular dystrophy (DMD). Although recent data suggest the role of H2S in ameliorating muscle wasting in murine and Caenorhabditis elegans models of DMD, possible cardioprotective effects have not yet been addressed. In this review, we summarize the current understanding of the role of H2S in animal models of cardiac dysfunctions and cardiac cells. We highlight that DMD may be amenable to H2S supplementation, and we suggest H2S as a possible factor regulating DMD-associated cardiomyopathy.
Collapse
Affiliation(s)
- Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7 Street, 30-387 Kraków, Poland;
| | | |
Collapse
|
7
|
Luo S, Kong C, Ye D, Liu X, Wang Y, Meng G, Han Y, Xie L, Ji Y. Protein Persulfidation: Recent Progress and Future Directions. Antioxid Redox Signal 2023; 39:829-852. [PMID: 36943282 DOI: 10.1089/ars.2022.0064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/23/2023]
Abstract
Significance: Hydrogen sulfide (H2S) is considered to be a gasotransmitter along with carbon monoxide (CO) and nitric oxide (NO), and is known as a key regulator of physiological and pathological activities. S-sulfhydration (also known as persulfidation), a mechanism involving the formation of protein persulfides by modification of cysteine residues, is proposed here to explain the multiple biological functions of H2S. Investigating the properties of protein persulfides can provide a foundation for further understanding of the potential functions of H2S. Recent Advances: Multiple methods have been developed to determine the level of protein persulfides. It has been demonstrated that protein persulfidation is involved in many biological processes through various mechanisms including the regulation of ion channels, enzymes, and transcription factors, as well as influencing protein-protein interactions. Critical Issues: Some technical and theoretical questions remain to be solved. These include how to improve the specificity of the detection methods for protein persulfidation, why persulfidation typically occurs on one or a few thiols within a protein, how this modification alters protein functions, and whether protein persulfidation has organ-specific patterns. Future Directions: Optimizing the detection methods and elucidating the properties and molecular functions of protein persulfidation would be beneficial for current therapeutics. In this review, we introduce the detailed mechanism of the persulfidation process and discuss persulfidation detection methods. In addition, this review summarizes recent discoveries of the selectivity of protein persulfidation and the regulation of protein functions and cell signaling pathways by persulfidation. Antioxid. Redox Signal. 39, 829-852.
Collapse
Affiliation(s)
- Shanshan Luo
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, China
| | - Chuiyu Kong
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, China
| | - Danyu Ye
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, China
| | - Xingeng Liu
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
| | - Yu Wang
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, China
| | - Guoliang Meng
- Department of Pharmacology, School of Pharmacy, Nantong University, Nantong, China
| | - Yi Han
- Department of Geriatrics, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Liping Xie
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
| | - Yong Ji
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Nanjing Medical University, Nanjing, China
- Key Laboratory of Targeted Intervention of Cardiovascular Disease, Nanjing Medical University, Nanjing, China
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, China
- The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Nanjing, China
- National Key Laboratory of Frigid Zone Cardiovascular Diseases (NKLFZCD), Harbin Medical University, Harbin, China
| |
Collapse
|
8
|
Piragine E, Citi V, Lawson K, Calderone V, Martelli A. Regulation of blood pressure by natural sulfur compounds: Focus on their mechanisms of action. Biochem Pharmacol 2022; 206:115302. [PMID: 36265595 DOI: 10.1016/j.bcp.2022.115302] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/10/2022] [Accepted: 10/11/2022] [Indexed: 12/14/2022]
Abstract
Natural sulfur compounds are emerging as therapeutic options for the management of hypertension and prehypertension. They are mainly represented by polysulfides from Alliaceae (i.e., garlic) and isothiocyanates from Brassicaceae (or crucifers). The beneficial cardiovascular effects of these compounds, especially garlic polysulfides, are well known and widely reported both in preclinical and clinical studies. However, only a few authors have linked the ability of natural sulfur compounds to induce vasorelaxation and subsequent antihypertensive effects with their ability to release hydrogen sulfide (H2S) in biological tissue. H2S is an endogenous gasotransmitter involved in vascular tone regulation. Some cardiovascular diseases, such as hypertension, are associated with lower plasma H2S levels. Consequently, exogenous sources of H2S (H2S donors) have been designed and synthesized or identified among secondary plant metabolites as potential therapeutic options. In addition to antioxidant effects due to its chemical properties as a reducing agent, H2S induces vasorelaxation by interacting with a range of molecular targets. The mechanisms of action accounting for H2S-induced vasodilation include opening of vascular potassium channels (such as ATP-sensitive (KATP) and voltage-operated (Kv7) channels), inhibition of 5-phosphodiesterase (5-PDE), and activation of vascular endothelial growth factor receptor-2 (VEGFR-2). These effects may be attributed to H2S-induced S-persulfidation (or S-sulfhydration), which is a posttranslational modification of cysteine residues of many types of proteins resulting in structural and functional alterations (activation/inhibition). Thus, H2S donors, such as natural sulfur compounds, are promising antihypertensive agents with novel mechanisms of action.
Collapse
Affiliation(s)
- Eugenia Piragine
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Valentina Citi
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy
| | - Kim Lawson
- Biomolecular Sciences Research Centre, Sheffield Hallam University, Sheffield S1 1WB, UK
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; Interdepartmental Research Center "Nutrafood: Nutraceutica e Alimentazione per la Salute", University of Pisa, 56126 Pisa, Italy; Interdepartmental Research Center "Biology and Pathology of Ageing", University of Pisa, 56126 Pisa, Italy
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, Via Bonanno 6, 56126 Pisa, Italy; Interdepartmental Research Center "Nutrafood: Nutraceutica e Alimentazione per la Salute", University of Pisa, 56126 Pisa, Italy; Interdepartmental Research Center "Biology and Pathology of Ageing", University of Pisa, 56126 Pisa, Italy.
| |
Collapse
|
9
|
Quan X, Chen W, Qin B, Wang J, Luo H, Dai F. The excitatory effect of hydrogen sulfide on rat colonic muscle contraction and the underlying mechanism. J Pharmacol Sci 2022; 149:100-107. [DOI: 10.1016/j.jphs.2022.04.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 03/29/2022] [Accepted: 04/13/2022] [Indexed: 10/18/2022] Open
|
10
|
Kaziród K, Myszka M, Dulak J, Łoboda A. Hydrogen sulfide as a therapeutic option for the treatment of Duchenne muscular dystrophy and other muscle-related diseases. Cell Mol Life Sci 2022; 79:608. [PMID: 36441348 PMCID: PMC9705465 DOI: 10.1007/s00018-022-04636-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 10/25/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022]
Abstract
Hydrogen sulfide (H2S) has been known for years as a poisoning gas and until recently evoked mostly negative associations. However, the discovery of its gasotransmitter functions suggested its contribution to various physiological and pathological processes. Although H2S has been found to exert cytoprotective effects through modulation of antioxidant, anti-inflammatory, anti-apoptotic, and pro-angiogenic responses in a variety of conditions, its role in the pathophysiology of skeletal muscles has not been broadly elucidated so far. The classical example of muscle-related disorders is Duchenne muscular dystrophy (DMD), the most common and severe type of muscular dystrophy. Mutations in the DMD gene that encodes dystrophin, a cytoskeletal protein that protects muscle fibers from contraction-induced damage, lead to prominent dysfunctions in the structure and functions of the skeletal muscle. However, the main cause of death is associated with cardiorespiratory failure, and DMD remains an incurable disease. Taking into account a wide range of physiological functions of H2S and recent literature data on its possible protective role in DMD, we focused on the description of the 'old' and 'new' functions of H2S, especially in muscle pathophysiology. Although the number of studies showing its essential regulatory action in dystrophic muscles is still limited, we propose that H2S-based therapy has the potential to attenuate the progression of DMD and other muscle-related disorders.
Collapse
Affiliation(s)
- Katarzyna Kaziród
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7, 30-387, Kraków, Poland
| | - Małgorzata Myszka
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7, 30-387, Kraków, Poland
| | - Józef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7, 30-387, Kraków, Poland
| | - Agnieszka Łoboda
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University in Krakow, Gronostajowa 7, 30-387, Kraków, Poland.
| |
Collapse
|
11
|
Li M, Mao J, Zhu Y. New Therapeutic Approaches Using Hydrogen Sulfide Donors in Inflammation and Immune Response. Antioxid Redox Signal 2021; 35:341-356. [PMID: 33789440 DOI: 10.1089/ars.2020.8249] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: Inflammation and immune response are associated with many pathological disorders, including rheumatoid arthritis, lupus, heart failure, and cancer(s). In recent times, important roles of hydrogen sulfide (H2S) have been evidenced by researchers in inflammatory responses, as well as immunomodulatory effects in several disease models. Recent Advances: Numerous biological targets, including cytochrome c oxidase, various kinases, enzymes involved in epigenetic changes, transcription factors, namely nuclear factor kappa B and nuclear factor erythroid 2-related factor 2, and several membrane ion channels, are shown to be sensitive to H2S and have been widely investigated in various preclinical models. Critical Issues: A complete understanding of the effects of H2S in inflammatory and immune response is vital in the development of novel H2S generating therapeutics. In this review, the biological effects and pharmacological properties of H2S in inflammation and immune response are addressed. The review also covers some of the novel H2S releasing prodrugs developed in recent years as tools to study this fascinating molecule. Future Directions: H2S plays important roles in inflammation and immunity-related processes. Future researches are needed to further assess the immunomodulatory effects of H2S and to assist in the design of more efficient H2S carrier systems, or drug formulations, for the management of immune-related conditions in humans. Antioxid. Redox Signal. 35, 341-356.
Collapse
Affiliation(s)
- Meng Li
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
| | - Jianchun Mao
- Department of Rheumatology, Longhua Hospital of Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yizhun Zhu
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, China
- School of Pharmacy, Macau University of Science and Technology, Macau, China
- Shanghai Key Laboratory of Bioactive Small Molecules, Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai, China
| |
Collapse
|
12
|
Hazzaa SM, Elsayed Arafat ESED, Abdo Ismail AEH, Eltorgoman AEA, Abdelaziz SA, Kombr YFA, Zidan RA, Assar MF. H 2S releasing Sodium sulfide protects from acute stress-induced hypertension by increasing the activity of endothelial nitric oxide synthase enzyme. Tissue Cell 2021; 72:101550. [PMID: 33915356 DOI: 10.1016/j.tice.2021.101550] [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: 02/27/2021] [Revised: 04/17/2021] [Accepted: 04/19/2021] [Indexed: 11/27/2022]
Abstract
Acute stress is a feature of our daily events that affects cardiovascular system and predisposes to hypertension. H2S is now considered as a vasorelaxant gasotransmitter although it was considered as a toxic agent. In present work we studied the effect of H2S releasing Na2S in acute stress induced hypertension and cardiac damage. Rats were divided into five groups: control, Na2S, acute stress, half dose of Na2S (6 mg/kg), and finally full dose of Na2S (12 mg/kg) to acute stressed rats. BP was measured then blood samples were taken for estimation of cortisol, cardiac enzymes markers, IL-6 and H2S. Finally, animals were sacrificed, hearts and thoracic aortae were excised for histological assessment, estimation of MDA, SOD and RNA extraction of CSE. Acute stress significantly elevated BP, cortisol, cardiac enzymes markers, IL-6, and tissue levels of MDA. It also, induced cardiac cell damage with congested B.V., extravasation of blood and decreased eNOs. Moreover, acute stress reduced H2S levels, RNA expression of CSE and SOD in cardiac tissues. Na2S significantly decreased BP, serum levels of cortisol, cardiac enzymes markers, IL-6, and tissue levels of MDA. Also, Na2S elevated serum H2S, RNA expression of CSE, SOD in cardiac tissue and increased eNOs activity.
Collapse
Affiliation(s)
- Suzan Moustafa Hazzaa
- Department of Medical Physiology, Faculty of Medicine, Menoufia University, Egypt; Department of Medical Physiology, Faculty of Medicine, Imam Mohammed Ibn Saud Islamic University, Saudi Arabia.
| | | | | | | | | | - Yasmin Fekry Abd Kombr
- Department of Chemistry, Biochemistry Division, Faculty of Science, Menoufia University, Egypt.
| | | | - Mohamed Farag Assar
- Department of Chemistry, Biochemistry Division, Faculty of Science, Menoufia University, Egypt.
| |
Collapse
|
13
|
Yang B, Zhao W, Yin C, Bai Y, Wang S, Xing G, Li F, Bian J, Aschner M, Cai J, Shi H, Lu R. Acute acrylonitrile exposure inhibits endogenous H 2S biosynthesis in rat brain and liver: The role of CBS/3-MPST-H 2S pathway in its astrocytic toxicity. Toxicology 2021; 451:152685. [PMID: 33486070 DOI: 10.1016/j.tox.2021.152685] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 12/12/2020] [Accepted: 01/15/2021] [Indexed: 12/11/2022]
Abstract
Hydrogen sulfide (H2S) as the third gasotransmitter molecule serves various biological regulatory roles in health and disease. Acrylonitrile (AN) is a common occupational toxicant and environmental pollutant, causing brain and liver damage in mammals. The biotransformation of AN is dependent-upon reduced glutathione (GSH), cysteine and other sulfur-containing compounds. However, the effects of AN on the endogenous H2S biosynthesis pathway have yet to be determined. Herein, we demonstrated that a single exposure to AN (at 25, 50, or 75 mg/kg for 1, 6 or 24 h) decreased the endogenous H2S content and H2S-producing capacity in a dose-dependent manner, both in the cerebral cortex and liver of rats in vivo. In addition, the inhibitory effects of AN (1, 2.5, 5, 10 mM for 12 h) on the H2S content and/or the expression of H2S-producing enzymes were also found both in primary rat astrocytes and rat liver cell line (BRL cells). Impairment in the H2S biosynthesis pathway was also assessed in primary rat astrocytes treated with AN. It was found that inhibition of the cystathionine-β-synthase (CBS)/3-mercaptopyruvate sulfurtransferase (3-MPST)-H2S pathway with the CBS inhibitor or 3-MPST-targeted siRNA significantly increased the AN-induced (5 mM for 12 h) cytotoxicity in astrocytes. In turn, CBS activation or 3-MPST overexpression as well as exogenous NaHS supplementation significantly attenuated AN-induced cytotoxicity. Taken together, endogenous H2S biosynthesis pathway was disrupted in rats acutely exposed to AN, which contributes to acute AN neurotoxicity in primary rat astrocytes.
Collapse
Affiliation(s)
- Bobo Yang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Wenjun Zhao
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China; Department of Clinical Laboratory, Affiliated People's Hospital to Jiangsu University School of Medicine, Zhenjiang, Jiangsu, 212002, China
| | - Changsheng Yin
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Yu Bai
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Suhua Wang
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Guangwei Xing
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Fang Li
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China
| | - Jinsong Bian
- Department of Pharmacology, School of Medicine, National Singapore University, 117597, Singapore
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, 10461, USA
| | - Jiyang Cai
- Department of Physiology, College of Medicine, University of Oklahoma Health Science Center, Lindsay, Oklahoma City, OK, 73104, USA
| | - Haifeng Shi
- School of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Rongzhu Lu
- Department of Preventive Medicine and Public Health Laboratory Science, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, Jiangsu, 212013, China; Center for Experimental Research, Affiliated Kunshan Hospital to Jiangsu University School of Medicine, Kunshan, Suzhou, Jiangsu, 215132, China.
| |
Collapse
|
14
|
Blachier F, Andriamihaja M, Larraufie P, Ahn E, Lan A, Kim E. Production of hydrogen sulfide by the intestinal microbiota and epithelial cells and consequences for the colonic and rectal mucosa. Am J Physiol Gastrointest Liver Physiol 2021; 320:G125-G135. [PMID: 33084401 DOI: 10.1152/ajpgi.00261.2020] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Among bacterial metabolites, hydrogen sulfide (H2S) has received increasing attention. The epithelial cells of the large intestine are exposed to two sources of H2S. The main one is the luminal source that results from specific bacteria metabolic activity toward sulfur-containing substrates. The other source in colonocytes is from the intracellular production mainly through cystathionine β-synthase (CBS) activity. H2S is oxidized by the mitochondrial sulfide oxidation unit, resulting in ATP synthesis, and, thus, establishing this compound as the first mineral energy substrate in colonocytes. However, when the intracellular H2S concentration exceeds the colonocyte capacity for its oxidation, it inhibits the mitochondrial respiratory chain, thus affecting energy metabolism. Higher luminal H2S concentration affects the integrity of the mucus layer and displays proinflammatory effects. However, a low/minimal amount of endogenous H2S exerts an anti-inflammatory effect on the colon mucosa, pointing out the ambivalent effect of H2S depending on its intracellular concentration. Regarding colorectal carcinogenesis, forced CBS expression in late adenoma-like colonocytes increased their proliferative activity, bioenergetics capacity, and tumorigenicity; whereas, genetic ablation of CBS in mice resulted in a reduced number of mutagen-induced aberrant crypt foci. Activation of endogenous H2S production and low H2S extracellular concentration enhance cancerous colorectal cell proliferation. Higher exogenous H2S concentrations markedly reduce mitochondrial ATP synthesis and proliferative capacity in cancerous cells and enhance glycolysis but do not affect their ATP cell content or viability. Thus, it appears that, notably through an effect on colonocyte energy metabolism, endogenous and microbiota-derived H2S are involved in the host intestinal physiology and physiopathology.
Collapse
Affiliation(s)
- François Blachier
- UMR PNCA, Nutrition Physiology and Alimentary Behavior, Université Paris-Saclay, AgroParisTech, INRAE, Paris, France
| | - Mireille Andriamihaja
- UMR PNCA, Nutrition Physiology and Alimentary Behavior, Université Paris-Saclay, AgroParisTech, INRAE, Paris, France
| | - Pierre Larraufie
- UMR PNCA, Nutrition Physiology and Alimentary Behavior, Université Paris-Saclay, AgroParisTech, INRAE, Paris, France
| | - Eunyeong Ahn
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan, South Korea
| | - Annaïg Lan
- UMR PNCA, Nutrition Physiology and Alimentary Behavior, Université Paris-Saclay, AgroParisTech, INRAE, Paris, France
| | - Eunjung Kim
- Department of Food Science and Nutrition, Daegu Catholic University, Gyeongsan, South Korea
| |
Collapse
|
15
|
Manandhar S, Sinha P, Ejiwale G, Bhatia M. Hydrogen Sulfide and its Interaction with Other Players in Inflammation. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1315:129-159. [PMID: 34302691 DOI: 10.1007/978-981-16-0991-6_6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hydrogen sulfide (H2S) plays a vital role in human physiology and in the pathophysiology of several diseases. In addition, a substantial role of H2S in inflammation has emerged. This chapter will discuss the involvement of H2S in various inflammatory diseases. Furthermore, the contribution of reactive oxygen species (ROS), adhesion molecules, and leukocyte recruitment in H2S-mediated inflammation will be discussed. The interrelationship of H2S with other gasotransmitters in inflammation will also be examined. There is mixed literature on the contribution of H2S to inflammation due to studies reporting both pro- and anti-inflammatory actions. These apparent discrepancies in the literature could be resolved with further studies.
Collapse
Affiliation(s)
- Sumeet Manandhar
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Priyanka Sinha
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Grace Ejiwale
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand.
| |
Collapse
|
16
|
Guerra DD, Hurt KJ. Gasotransmitters in pregnancy: from conception to uterine involution. Biol Reprod 2020; 101:4-25. [PMID: 30848786 DOI: 10.1093/biolre/ioz038] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 02/14/2019] [Accepted: 03/06/2019] [Indexed: 12/13/2022] Open
Abstract
Gasotransmitters are endogenous small gaseous messengers exemplified by nitric oxide (NO), carbon monoxide (CO), and hydrogen sulfide (H2S or sulfide). Gasotransmitters are implicated in myriad physiologic functions including many aspects of reproduction. Our objective was to comprehensively review basic mechanisms and functions of gasotransmitters during pregnancy from conception to uterine involution and highlight future research opportunities. We searched PubMed and Web of Science databases using combinations of keywords nitric oxide, carbon monoxide, sulfide, placenta, uterus, labor, and pregnancy. We included English language publications on human and animal studies from any date through August 2018 and retained basic and translational articles with relevant original findings. All gasotransmitters activate cGMP signaling. NO and sulfide also covalently modify target protein cysteines. Protein kinases and ion channels transduce gasotransmitter signals, and co-expressed gasotransmitters can be synergistic or antagonistic depending on cell type. Gasotransmitters influence tubal transit, placentation, cervical remodeling, and myometrial contractility. NO, CO, and sulfide dilate resistance vessels, suppress inflammation, and relax myometrium to promote uterine quiescence and normal placentation. Cervical remodeling and rupture of fetal membranes coincide with enhanced oxidation and altered gasotransmitter metabolism. Mechanisms mediating cellular and organismal changes in pregnancy due to gasotransmitters are largely unknown. Altered gasotransmitter signaling has been reported for preeclampsia, intrauterine growth restriction, premature rupture of membranes, and preterm labor. However, in most cases specific molecular changes are not yet characterized. Nonclassical signaling pathways and the crosstalk among gasotransmitters are emerging investigation topics.
Collapse
Affiliation(s)
- Damian D Guerra
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| | - K Joseph Hurt
- Division of Reproductive Sciences, Department of Obstetrics and Gynecology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA.,Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| |
Collapse
|
17
|
An Indole Alkaloid Extracted from Evodia rutaecarpa Inhibits Colonic Motility of Rats In Vitro. Gastroenterol Res Pract 2020; 2020:8610653. [PMID: 32328100 PMCID: PMC7157783 DOI: 10.1155/2020/8610653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/14/2020] [Accepted: 03/18/2020] [Indexed: 11/17/2022] Open
Abstract
Evodiamine (Evo) is an indole alkaloid extracted from the traditional Chinese medicinal herb Evodia rutaecarpa. Evo may regulate gastrointestinal motility, but the evidence is insufficient, and the mechanisms remain unknown. The aim of this study was to investigate the effect of Evo on colonic motility of rats and the underlying mechanisms in vitro. Rat colonic muscle was exposed to Evo (10 and 100 μM) followed by immunohistochemistry of cholecystokinin receptor 1 (CCK1R). Muscle contractions were studied in an organ bath system to determine whether CCK1R, nitric oxide (NO), and enteric neurons are involved in the relaxant effect of Evo. Whole-cell patch-clamp was used to detect L-type calcium currents (ICa,L) in isolated colonic smooth muscle cells (SMCs). CCK1R was observed in SMCs, intermuscular neurons, and mucosa of rat colon. Evo could inhibit spontaneous muscle contractions; NO synthase, inhibitor L-NAME CCK1R antagonist, could partly block this effect, while the enteric neurons may not play a major role. Evo inhibited the peak ICa,L in colonic SMCs at a membrane potential of 0 mV. The current-voltage (I–V) relationship of L-type calcium channels was modified by Evo, while the peak of the I–V curve remained at 0 mV. Furthermore, Evo inhibited the activation of L-type calcium channels and decreased the peak ICa,L. The relaxant effect of Evo on colonic muscle is associated with the inhibition of L-type calcium channels. The enteric neurons, NO, and CCK1R may be partly related to the inhibitory effect of Evo on colonic motility. This study provides the first evidence that evodiamine can regulate colonic motility in rats by mediating calcium homeostasis in smooth muscle cells. These data form a theoretical basis for the clinical application of evodiamine for treatment of gastrointestinal motility diseases.
Collapse
|
18
|
Tabassum R, Jeong NY, Jung J. Therapeutic importance of hydrogen sulfide in age-associated neurodegenerative diseases. Neural Regen Res 2020; 15:653-662. [PMID: 31638087 PMCID: PMC6975154 DOI: 10.4103/1673-5374.266911] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 04/27/2019] [Accepted: 08/01/2019] [Indexed: 02/06/2023] Open
Abstract
Hydrogen sulfide (H2S) is a gasotransmitter that acts as an antioxidant and exhibits a wide variety of cytoprotective and physiological functions in age-associated diseases. One of the major causes of age-related diseases is oxidative stress. In recent years, the importance of H2S has become clear, although its antioxidant function has not yet been fully explored. The enzymes cystathionine β-synthase, cystathionine γ-lya-se, and 3-mercaptopyruvate sulfurtransferase are involved in the enzymatic production of H2S. Previously, H2S was considered a neuromodulator, given its role in long-term hippocampal potentiation, but it is now also recognized as an antioxidant in age-related neurodegeneration. Due to aerobic metabolism, the central nervous system is vulnerable to oxidative stress in brain aging, resulting in age-associated degenerative diseases. H2S exerts its antioxidant effect by limiting free radical reactions through the activation of antioxidant enzymes, including superoxide dismutase, catalase, and glutathione peroxidase, which protect against the effects of aging by regulating apoptosis-related genes, including p53, Bax, and Bcl-2. This review explores the implications and mechanisms of H2S as an antioxidant in age-associated neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Down syndrome.
Collapse
Affiliation(s)
- Rubaiya Tabassum
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan, Korea
- Department of Medicine, Graduate School, Dong-A University, Busan, Korea
| | - Na Young Jeong
- Department of Anatomy and Cell Biology, College of Medicine, Dong-A University, Busan, Korea
- Department of Medicine, Graduate School, Dong-A University, Busan, Korea
| | - Junyang Jung
- Department of Anatomy and Neurobiology, College of Medicine, Kyung Hee University, Seoul, Korea
| |
Collapse
|
19
|
Wetzel MD, Wenke JC. Mechanisms by which hydrogen sulfide attenuates muscle function following ischemia-reperfusion injury: effects on Akt signaling, mitochondrial function, and apoptosis. J Transl Med 2019; 17:33. [PMID: 30665344 PMCID: PMC6340183 DOI: 10.1186/s12967-018-1753-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2018] [Accepted: 12/21/2018] [Indexed: 01/24/2023] Open
Abstract
Ischemia–reperfusion injury is caused by a period of ischemia followed by massive blood flow into a tissue that had experienced restricted blood flow. The severity of the injury is dependent on the time the tissue was restricted from blood flow, becoming more severe after longer ischemia times. This can lead to many complications such as tissue necrosis, cellular apoptosis, inflammation, metabolic and mitochondrial dysfunction, and even organ failure. One of the emerging therapies to combat ischemic reperfusion injury complications is hydrogen sulfide, which is a gasotransmitter that diffuses across cell membranes to exert effects on various signaling pathways regulating cell survival such as Akt, mitochondrial activity, and apoptosis. Although commonly thought of as a toxic gas, low concentrations of hydrogen sulfide have been shown to be beneficial in promoting tissue survival post-ischemia, and modulate a wide variety of cellular responses. This review will detail the mechanisms of hydrogen sulfide in affecting the Akt signaling pathway, mitochondrial function, and apoptosis, particularly in regards to ischemic reperfusion injury in muscle tissue. It will conclude with potential clinical applications of hydrogen sulfide, combinations with other therapies, and perspectives for future studies.
Collapse
Affiliation(s)
- Michael D Wetzel
- US Army Institute of Surgical Research, Extremity Trauma and Regenerative Medicine, 3698 Chambers Pass BLDG 3611, Ft. Sam Houston, San Antonio, TX, 78234, USA
| | - Joseph C Wenke
- US Army Institute of Surgical Research, Extremity Trauma and Regenerative Medicine, 3698 Chambers Pass BLDG 3611, Ft. Sam Houston, San Antonio, TX, 78234, USA.
| |
Collapse
|
20
|
Nalli AD, Wang H, Bhattacharya S, Blakeney BA, Murthy KS. Inhibition of RhoA/Rho kinase pathway and smooth muscle contraction by hydrogen sulfide. Pharmacol Res Perspect 2018; 5. [PMID: 28971603 PMCID: PMC5625153 DOI: 10.1002/prp2.343] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 12/22/2022] Open
Abstract
Hydrogen sulfide (H2 S) plays an important role in smooth muscle relaxation. Here, we investigated the expression of enzymes in H2 S synthesis and the mechanism regulating colonic smooth muscle function by H2 S. Expression of cystathionine-γ-lyase (CSE), but not cystathionine-β-synthase (CBS), was identified in the colonic smooth muscle of rabbit, mouse, and human. Carbachol (CCh)-induced contraction in rabbit muscle strips and isolated muscle cells was inhibited by l-cysteine (substrate of CSE) and NaHS (an exogenous H2 S donor) in a concentration-dependent fashion. H2 S induced S-sulfhydration of RhoA that was associated with inhibition of RhoA activity. CCh-induced Rho kinase activity also was inhibited by l-cysteine and NaHS in a concentration-dependent fashion. Inhibition of CCh-induced contraction by l-cysteine was blocked by the CSE inhibitor, dl-propargylglycine (DL-PPG) in dispersed muscle cells. Inhibition of CCh-induced Rho kinase activity by l-cysteine was blocked by CSE siRNA in cultured cells and DL-PPG in dispersed muscle cells. Stimulation of Rho kinase activity and muscle contraction in response to CCh was also inhibited by l-cysteine or NaHS in colonic muscle cells from mouse and human. Collectively, our studies identified the expression of CSE in colonic smooth muscle and determined that sulfhydration of RhoA by H2 S leads to inhibition of RhoA and Rho kinase activities and muscle contraction. The mechanism identified may provide novel therapeutic approaches to mitigate gastrointestinal motility disorders.
Collapse
Affiliation(s)
- Ancy D Nalli
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Hongxia Wang
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Sayak Bhattacharya
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Bryan A Blakeney
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Karnam S Murthy
- Department of Physiology and Biophysics, VCU Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
21
|
Ha J, Xu Y, Kawano T, Hendon T, Baki L, Garai S, Papapetropoulos A, Thakur GA, Plant LD, Logothetis DE. Hydrogen sulfide inhibits Kir2 and Kir3 channels by decreasing sensitivity to the phospholipid phosphatidylinositol 4,5-bisphosphate (PIP 2). J Biol Chem 2018; 293:3546-3561. [PMID: 29317494 DOI: 10.1074/jbc.ra117.001679] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 01/07/2018] [Indexed: 11/06/2022] Open
Abstract
Inwardly rectifying potassium (Kir) channels establish and regulate the resting membrane potential of excitable cells in the heart, brain, and other peripheral tissues. Phosphatidylinositol 4,5-bisphosphate (PIP2) is a key direct activator of ion channels, including Kir channels. The gasotransmitter carbon monoxide has been shown to regulate Kir channel activity by altering channel-PIP2 interactions. Here, we tested in two cellular models the effects and mechanism of action of another gasotransmitter, hydrogen sulfide (H2S), thought to play a key role in cellular responses under ischemic conditions. Direct administration of sodium hydrogen sulfide as an exogenous H2S source and expression of cystathionine γ-lyase, a key enzyme that produces endogenous H2S in specific brain tissues, resulted in comparable current inhibition of several Kir2 and Kir3 channels. This effect resulted from changes in channel-gating kinetics rather than in conductance or cell-surface localization. The extent of H2S regulation depended on the strength of the channel-PIP2 interactions. H2S regulation was attenuated when channel-PIP2 interactions were strengthened and was increased when channel-PIP2 interactions were weakened by depleting PIP2 levels. These H2S effects required specific cytoplasmic cysteine residues in Kir3.2 channels. Mutation of these residues abolished H2S inhibition, and reintroduction of specific cysteine residues back into the background of the cytoplasmic cysteine-lacking mutant rescued H2S inhibition. Molecular dynamics simulation experiments provided mechanistic insights into how potential sulfhydration of specific cysteine residues could lead to changes in channel-PIP2 interactions and channel gating.
Collapse
Affiliation(s)
- Junghoon Ha
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Yu Xu
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Takeharu Kawano
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Tyler Hendon
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298
| | - Lia Baki
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Sumanta Garai
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Andreas Papapetropoulos
- the Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens 157 71, Greece, and.,the Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens 11527, Greece
| | - Ganesh A Thakur
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Leigh D Plant
- Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| | - Diomedes E Logothetis
- From the Department of Physiology and Biophysics, Virginia Commonwealth University School of Medicine, Richmond, Virginia 23298, .,Department of Pharmaceutical Sciences in the School of Pharmacy, Northeastern University Bouvé College of Health Sciences, Boston, Massachusetts 02115
| |
Collapse
|
22
|
Lesniak A, Aarnio M, Diwakarla S, Norberg T, Nyberg F, Gordh T. Characterization of the binding site for d-deprenyl in human inflamed synovial membrane. Life Sci 2017; 194:26-33. [PMID: 29221756 DOI: 10.1016/j.lfs.2017.12.003] [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] [Received: 09/01/2017] [Revised: 11/28/2017] [Accepted: 12/03/2017] [Indexed: 01/23/2023]
Abstract
AIMS d-Deprenyl when used as a positron emission tomography tracer visualizes peripheral inflammation. The major aim of the current study was to identify and investigate the properties of the binding target for d-deprenyl in synovial membrane explants from arthritic patients. MAIN METHODS Thirty patients diagnosed with arthritis or osteoarthritis were enrolled into the study. Homologous and competitive radioligand binding assays utilizing [3H]d-deprenyl were performed to investigate the biochemical characteristics of the binding site and assess differences in the binding profile in synovial membranes exhibiting varying levels of inflammation. KEY FINDINGS The [3H]d-deprenyl binding assay confirmed the existence of a single, saturable population of membrane-bound protein binding sites in synovial membrane homogenates. The macroscopically determined level of inflammation correlated with an increase in [3H]d-deprenyl binding affinity, without significant alterations in binding site density. Selective monoamine oxidase B inhibitor, selegiline competed for the same site as [3H]d-deprenyl, but failed to differentiate the samples with regard to their inflammation grade. A monoamine oxidase A inhibitor, pirlindole mesylate showed only weak displacement of [3H]d-deprenyl binding. No significant alterations in monoamine oxidase B expression was detected, thus it was not confirmed whether it could serve as a marker for ongoing inflammation. SIGNIFICANCE Our study was the first to show the biochemical characteristics of the [3H]d-deprenyl binding site in inflamed human synovium. We confirmed that d-deprenyl could differentiate between patients with varying severity of synovitis in the knee joint by binding to a protein target distinct from monoamine oxidase B.
Collapse
Affiliation(s)
- Anna Lesniak
- Uppsala University, Department of Pharmaceutical Biosciences, SE 751 24 Uppsala, Sweden; Medical University of Warsaw, Department of Pharmacodynamics, Centre for Preclinical Research and Technology, 02-097 Warsaw, Poland.
| | - Mikko Aarnio
- Uppsala University Hospital, Department of Surgical Sciences, Anaesthesiology and Intensive Care, SE 751 85 Uppsala, Sweden
| | - Shanti Diwakarla
- Uppsala University, Department of Pharmaceutical Biosciences, SE 751 24 Uppsala, Sweden
| | - Thomas Norberg
- Uppsala University, Department of Physical Organic Chemistry, SE 751 23 Uppsala, Sweden
| | - Fred Nyberg
- Uppsala University, Department of Pharmaceutical Biosciences, SE 751 24 Uppsala, Sweden
| | - Torsten Gordh
- Uppsala University Hospital, Department of Surgical Sciences, Anaesthesiology and Intensive Care, SE 751 85 Uppsala, Sweden
| |
Collapse
|
23
|
Nalli AD, Bhattacharya S, Wang H, Kendig DM, Grider JR, Murthy KS. Augmentation of cGMP/PKG pathway and colonic motility by hydrogen sulfide. Am J Physiol Gastrointest Liver Physiol 2017; 313:G330-G341. [PMID: 28705807 PMCID: PMC5668569 DOI: 10.1152/ajpgi.00161.2017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 06/26/2017] [Accepted: 06/27/2017] [Indexed: 01/31/2023]
Abstract
Hydrogen sulfide (H2S), like nitric oxide (NO), causes smooth muscle relaxation, but unlike NO, does not stimulate soluble guanylyl cyclase (sGC) activity and generate cyclic guanosine 5'-monophosphate (cGMP). The aim of this study was to investigate the interplay between NO and H2S in colonic smooth muscle. In colonic smooth muscle from rabbit, mouse, and human, l-cysteine, substrate of cystathionine-γ-lyase (CSE), or NaHS, an H2S donor, inhibited phosphodiesterase 5 (PDE5) activity and augmented the increase in cGMP levels, IP3 receptor phosphorylation at Ser1756 (measured as a proxy for PKG activation), and muscle relaxation in response to NO donor S-nitrosoglutathione (GSNO), suggesting augmentation of cGMP/PKG pathway by H2S. The inhibitory effect of l-cysteine, but not NaHS, on PDE5 activity was blocked in cells transfected with CSE siRNA or treated with CSE inhibitor d,l-propargylglycine (dl-PPG), suggesting activation of CSE and generation of H2S in response to l-cysteine. H2S levels were increased in response to l-cysteine, and the effect of l-cysteine was augmented by GSNO in a cGMP-dependent protein kinase-sensitive manner, suggesting augmentation of CSE/H2S by cGMP/PKG pathway. As a result, GSNO-induced relaxation was inhibited by dl-PPG. In flat-sheet preparation of colon, l-cysteine augmented calcitonin gene-related peptide release in response to mucosal stimulation, and in intact segments, l-cysteine increased the velocity of pellet propulsion. These results demonstrate that in colonic smooth muscle, there is a novel interplay between NO and H2S. NO generates H2S via cGMP/PKG pathway, and H2S, in turn, inhibits PDE5 activity and augments NO-induced cGMP levels. In the intact colon, H2S promotes colonic transit.NEW & NOTEWORTHY Hydrogen sulfide (H2S) and nitric oxide (NO) are important regulators of gastrointestinal motility. The studies herein provide the cross talk between NO and H2S signaling to mediate smooth muscle relaxation and colonic transit. H2S inhibits phosphodiesterase 5 activity to augment cGMP levels in response to NO, which, in turn, via cGMP/PKG pathway, generates H2S. These studies suggest that interventions targeted at restoring NO and H2S homeostasis within the smooth muscle may provide novel therapeutic approaches to mitigate motility disorders.
Collapse
Affiliation(s)
- Ancy D Nalli
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Sayak Bhattacharya
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Hongxia Wang
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Derek M Kendig
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - John R Grider
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Karnam S Murthy
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
24
|
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
|
25
|
Kanagy NL, Szabo C, Papapetropoulos A. Vascular biology of hydrogen sulfide. Am J Physiol Cell Physiol 2017; 312:C537-C549. [PMID: 28148499 PMCID: PMC5451519 DOI: 10.1152/ajpcell.00329.2016] [Citation(s) in RCA: 156] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 01/24/2017] [Accepted: 01/27/2017] [Indexed: 12/23/2022]
Abstract
Hydrogen sulfide (H2S) is a ubiquitous signaling molecule with important functions in many mammalian organs and systems. Observations in the 1990s ascribed physiological actions to H2S in the nervous system, proposing that this gasotransmitter acts as a neuromodulator. Soon after that, the vasodilating properties of H2S were demonstrated. In the past decade, H2S was shown to exert a multitude of physiological effects in the vessel wall. H2S is produced by vascular cells and exhibits antioxidant, antiapoptotic, anti-inflammatory, and vasoactive properties. In this concise review, we have focused on the impact of H2S on vascular structure and function with an emphasis on angiogenesis, vascular tone, vascular permeability and atherosclerosis. H2S reduces arterial blood pressure, limits atheromatous plaque formation, and promotes vascularization of ischemic tissues. Although the beneficial properties of H2S are well established, mechanistic insights into the molecular pathways implicated in disease prevention and treatment remain largely unexplored. Unraveling the targets and downstream effectors of H2S in the vessel wall in the context of disease will aid in translation of preclinical observations. In addition, acute regulation of H2S production is still poorly understood and additional work delineating the pathways regulating the enzymes that produce H2S will allow pharmacological manipulation of this pathway. As the field continues to grow, we expect that H2S-related compounds will find their way into clinical trials for diseases affecting the blood vessels.
Collapse
Affiliation(s)
- Nancy L Kanagy
- Vascular Physiology Group, Department of Cell Biology and Physiology, School of Medicine, University of New Mexico, Albuquerque, New Mexico
| | - Csaba Szabo
- Department of Anesthesiology, University of Texas Medical Branch, Galveston, Texas
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece; and
- Center of Clinical, Experimental Surgery and Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| |
Collapse
|
26
|
H2S-induced thiol-based redox switches: Biochemistry and functional relevance for inflammatory diseases. Pharmacol Res 2016; 111:642-651. [PMID: 27468648 DOI: 10.1016/j.phrs.2016.07.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 07/19/2016] [Accepted: 07/22/2016] [Indexed: 02/08/2023]
Abstract
During the last decades, small inorganic molecules like reactive oxygen species (ROS), nitric oxide (NO), carbon monoxide (CO) and even the highly toxic hydrogen sulfide (H2S) have been evolved as important signaling molecules that trigger crucial cellular processes by regulating the activity of kinases, phosphatases and transcription factors. These redox molecules use similar target structures and therefore, the composition of the complex "redox environment" determines the final outcome of signaling processes and may subsequently also affect the behavior of a cell in an inflammatory environment. Here, we discuss the role of H2S in this complex interplay with a focus on the transcription factors Nrf2 and NFκB.
Collapse
|
27
|
Abstract
Voltage-gated ion channels are key regulators of cell excitability. There is significant evidence that these channels are subject to modulation by redox status of the cells. Here we review the post-translational modifications of ion channels that occur in colonic inflammation. The redox mechanisms involve tyrosine nitration, covalent modification of cysteine residues and sulfhydration by hydrogen sulfide in experimental colitis. In the setting of colonic inflammation, modifications of cysteine and tyrosine are likely to occur at several sites within the same channel complex. In this review we describe alterations in channel function due to specific modifications of tyrosine and cysteine residues by reactive nitrogen, oxygen and hydrogen-sulfide resulting in altered motility.
Collapse
Affiliation(s)
- Hamid I Akbarali
- Department of Pharmacology and Toxicology, Virginia Commonwealth University, 1112 East Clay Street, McGuire Hall Rm# 317, Richmond, VA 23298, USA Country.
| | | |
Collapse
|
28
|
Li B, Zani A, Martin Z, Lee C, Zani-Ruttenstock E, Eaton S, Pierro A. Intestinal epithelial cell injury is rescued by hydrogen sulfide. J Pediatr Surg 2016; 51:775-8. [PMID: 26947403 DOI: 10.1016/j.jpedsurg.2016.02.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 02/07/2016] [Indexed: 12/22/2022]
Abstract
BACKGROUND/PURPOSE Oxidative stress is implicated in the pathogenesis of necrotizing enterocolitis (NEC). Hydrogen sulfide (H2S) has been reported to have a protective function against oxidative stress in the gut. We hypothesize that administration of H2S can help decrease intestinal epithelial cell injury in vitro. METHODS Intestinal epithelial cells (IEC-18) were treated with 200μM hydrogen peroxide (H2O2) for 21h. At 21h sodium hydrosulfide (NaHS), an H2S donor, was administered as a rescue treatment at two different concentrations: 0.1mM and 0.2mM. At 24h, cell viability was measured using a colorimetric assay (MTT). Oxidative stress was studied by glutathione peroxidase (GPx) activity and thiobarbituric acid reactive substances (TBARS). IL-6 and TNFα levels were tested to study inflammation. Data were presented as mean±SD and compared using one-way ANOVA with Bonferroni post-test. RESULTS Compared to control, H2O2-treated IEC-18 had reduced viability (p<0.01), lower GPx activity (p<0.01), higher TBARS levels (p<0.01), and increased IL6 and TNFα (p<0.001). Compared to H2O2-treated IEC-18, treatment with 0.2mM NaHS rescued viability (p<0.01), increased GPx activity (p<0.05), and reduced TBARS (p<0.01), IL6 and TNFα (p<0.001). CONCLUSIONS H2S successfully rescues epithelial cell damage induced by oxidative stress in vitro. This indicates that H2S could be a potential pharmacological intervention in conditions like NEC.
Collapse
Affiliation(s)
- Bo Li
- Division of General and Thoracic Surgery, Physiology and Experimental Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Augusto Zani
- Division of General and Thoracic Surgery, Physiology and Experimental Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Zechariah Martin
- Division of General and Thoracic Surgery, Physiology and Experimental Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Carol Lee
- Division of General and Thoracic Surgery, Physiology and Experimental Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Elke Zani-Ruttenstock
- Division of General and Thoracic Surgery, Physiology and Experimental Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada
| | - Simon Eaton
- UCL Institute of Child Health, London, United Kingdom
| | - Agostino Pierro
- Division of General and Thoracic Surgery, Physiology and Experimental Medicine Program, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
29
|
Magalhães D, Cabral JM, Soares-da-Silva P, Magro F. Role of epithelial ion transports in inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol 2016; 310:G460-76. [PMID: 26744474 DOI: 10.1152/ajpgi.00369.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 01/02/2016] [Indexed: 02/06/2023]
Abstract
Inflammatory bowel disease (IBD) is a chronic inflammatory disorder with a complex pathogenesis. Diarrhea is a highly prevalent and often debilitating symptom of IBD patients that results, at least in part, from an intestinal hydroelectrolytic imbalance. Evidence suggests that reduced electrolyte absorption is more relevant than increased secretion to this disequilibrium. This systematic review analyses and integrates the current evidence on the roles of epithelial Na(+)-K(+)-ATPase (NKA), Na(+)/H(+) exchangers (NHEs), epithelial Na(+) channels (ENaC), and K(+) channels (KC) in IBD-associated diarrhea. NKA is the key driving force of the transepithelial ionic transport and its activity is decreased in IBD. In addition, the downregulation of apical NHE and ENaC and the upregulation of apical large-conductance KC all contribute to the IBD-associated diarrhea by lowering sodium absorption and/or increasing potassium secretion.
Collapse
Affiliation(s)
- Diogo Magalhães
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Porto, Portugal; and MedInUP-Center for Drug Discovery and Innovative Medicines, University of Porto, Porto, Portugal
| | - José Miguel Cabral
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Porto, Portugal; and MedInUP-Center for Drug Discovery and Innovative Medicines, University of Porto, Porto, Portugal
| | - Patrício Soares-da-Silva
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Porto, Portugal; and MedInUP-Center for Drug Discovery and Innovative Medicines, University of Porto, Porto, Portugal
| | - Fernando Magro
- Department of Pharmacology and Therapeutics, Faculty of Medicine, Porto, Portugal; and MedInUP-Center for Drug Discovery and Innovative Medicines, University of Porto, Porto, Portugal
| |
Collapse
|
30
|
Hydrogen sulfide diminishes the levels of thymic stromal lymphopoietin in activated mast cells. Arch Dermatol Res 2016; 308:103-13. [DOI: 10.1007/s00403-016-1619-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 09/14/2015] [Accepted: 01/08/2016] [Indexed: 12/30/2022]
|
31
|
Abstract
This review is focused on formation and biological significance of hydropersulfides, i.e. S-sulfhydration process. Biogenesis and properties of reactive sulfur species and their role in redox signaling are presented. The effect of S-sulfhydration on protein function is discussed. For many years reactive oxygen and nitrogen species (ROS and RNS) have been recognized as key messengers in the process of thiol-based redox regulation. Relatively recently, literature reports began to mention reactive sulfur species (RSS) and their role in thiol regulation. This review is focused on biogenesis and biological properties of RSS, including: hydropersulfides, polysulfides and hydrogen sulfide (H2S). Based on the most up-to-date literature data, the paper presents biological significance of S-sulfhydration process. In this reaction, sulfane sulfur is transferred to the–SH groups forming hydropersulfides. Protein cysteine residues, called ‘redox switches’ are susceptible to such reversible modifications. In line with the most recent reports, it was emphasized that sulfane sulfur-containing compounds (mainly hydrogen persulfides and polysulfides) are real and better mediators of S-sulfhydration-based signalling than H2S. We also overviewed proteins participating in the formation and transport of RSS and in mitochondrial H2S oxidation. In addition, we reviewed many reports about proteins unrelated to sulfur metabolism which are modified by S-sulfhydration that influences their catalytic activity. We also addressed the problem of the regulatory function of S-sulfhydration reaction in the activation of KATP channels (vasorelaxant) and transcription factors (e.g. NFκB) as well as in the mechanism of therapeutic action of garlic-derived sulfur compounds. Some aspects of comparison between RNS and RSS are also discussed in this review.
Collapse
|
32
|
Nelson PT, Jicha GA, Wang WX, Ighodaro E, Artiushin S, Nichols CG, Fardo DW. ABCC9/SUR2 in the brain: Implications for hippocampal sclerosis of aging and a potential therapeutic target. Ageing Res Rev 2015; 24:111-25. [PMID: 26226329 PMCID: PMC4661124 DOI: 10.1016/j.arr.2015.07.007] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 07/24/2015] [Indexed: 01/06/2023]
Abstract
The ABCC9 gene and its polypeptide product, SUR2, are increasingly implicated in human neurologic disease, including prevalent diseases of the aged brain. SUR2 proteins are a component of the ATP-sensitive potassium ("KATP") channel, a metabolic sensor for stress and/or hypoxia that has been shown to change in aging. The KATP channel also helps regulate the neurovascular unit. Most brain cell types express SUR2, including neurons, astrocytes, oligodendrocytes, microglia, vascular smooth muscle, pericytes, and endothelial cells. Thus it is not surprising that ABCC9 gene variants are associated with risk for human brain diseases. For example, Cantu syndrome is a result of ABCC9 mutations; we discuss neurologic manifestations of this genetic syndrome. More common brain disorders linked to ABCC9 gene variants include hippocampal sclerosis of aging (HS-Aging), sleep disorders, and depression. HS-Aging is a prevalent neurological disease with pathologic features of both neurodegenerative (aberrant TDP-43) and cerebrovascular (arteriolosclerosis) disease. As to potential therapeutic intervention, the human pharmacopeia features both SUR2 agonists and antagonists, so ABCC9/SUR2 may provide a "druggable target", relevant perhaps to both HS-Aging and Alzheimer's disease. We conclude that more work is required to better understand the roles of ABCC9/SUR2 in the human brain during health and disease conditions.
Collapse
Affiliation(s)
- Peter T Nelson
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA; University of Kentucky, Department of Pathology, Lexington, KY 40536, USA.
| | - Gregory A Jicha
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA; University of Kentucky, Department of Neurology, Lexington, KY, 40536, USA
| | - Wang-Xia Wang
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA
| | - Eseosa Ighodaro
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA
| | - Sergey Artiushin
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA
| | - Colin G Nichols
- Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | - David W Fardo
- University of Kentucky, Sanders-Brown Center on Aging, Lexington, KY 40536, USA; Department of Biostatistics, Lexington, KY, 40536, USA
| |
Collapse
|
33
|
Hydrogen sulfide-based therapeutics: exploiting a unique but ubiquitous gasotransmitter. Nat Rev Drug Discov 2015; 14:329-45. [PMID: 25849904 DOI: 10.1038/nrd4433] [Citation(s) in RCA: 602] [Impact Index Per Article: 60.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Hydrogen sulfide (H2S) has become recognized as an important signalling molecule throughout the body, contributing to many physiological and pathological processes. In recent years, improved methods for measuring H2S levels and the availability of a wider range of H2S donors and more selective inhibitors of H2S synthesis have helped to more accurately identify the many biological effects of this highly reactive gaseous mediator. Animal studies of several H2S-releasing drugs have demonstrated considerable promise for the safe treatment of a wide range of disorders. Several such drugs are now in clinical trials.
Collapse
|
34
|
Quan X, Luo H, Liu Y, Xia H, Chen W, Tang Q. Hydrogen sulfide regulates the colonic motility by inhibiting both L-type calcium channels and BKCa channels in smooth muscle cells of rat colon. PLoS One 2015; 10:e0121331. [PMID: 25811907 PMCID: PMC4374679 DOI: 10.1371/journal.pone.0121331] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2014] [Accepted: 01/30/2015] [Indexed: 12/18/2022] Open
Abstract
Objective To examine the hypothesis that hydrogen sulfide (H2S) regulates the colonic motility by modulating both L-type voltage-dependent calcium channels and large conductance Ca2+-activated K+ (BKCa) channels. Methods Immunohistochemistry was performed on rat colonic samples to investigate the localization of the H2S-producing enzymes cystathionine-β-synthase (CBS) and cystathionine-γ-lyase (CSE). The contractions of proximal colonic smooth muscle were studied in an organ bath system. The whole-cell patch-clamp technique was used to record both L-type calcium currents (ICa,L) and BKCa currents in colonic smooth muscle cells (SMCs) isolated from male Wistar rats. Results Immunohistochemistry revealed the presence of CBS and CSE in mucosa, smooth muscle cells and myenteric neurons. The H2S donor NaHS inhibited spontaneous contractions of the longitudinal muscle and circular muscle strips in a dose-dependent manner, and the inhibitory effects were not blocked by tetrodotoxin. NaHS inhibited the peak ICa,L in colonic SMCs at a membrane potential of 0 mV. The current-voltage (I-V) relationship of L-type calcium channels was modified by NaHS, and the peak of the I-V curve was shifted to the right. NaHS (200μΜ) evoked a significant rightward shift of the steady-state activation curve and inhibited the inactivation of L-type calcium channels. Furthermore, NaHS reversibly decreased the peak ICa,L in a dose-dependent manner. Likewise, BKCa channels were significantly inhibited by NaHS, and the addition of NaHS caused a time- and dose-dependent reduction in the BKCa current. Conclusion The relaxant effect of H2S on colonic muscle strips may be associated with the direct inhibition of H2S on L-type calcium channels. H2S may be involved in the regulation of calcium homeostasis in colonic SMCs of rat colon.
Collapse
Affiliation(s)
- Xiaojing Quan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Hesheng Luo
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- * E-mail:
| | - Yin Liu
- Department of Gastroenterology, the Affiliated Hospital of Guilin Medical College, Guilin, China
| | - Hong Xia
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
- Key Laboratory of Hubei Province for Digestive System Diseases, Wuhan, China
| | - Wei Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qincai Tang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
35
|
Kang M, Hashimoto A, Gade A, Akbarali HI. Interaction between hydrogen sulfide-induced sulfhydration and tyrosine nitration in the KATP channel complex. Am J Physiol Gastrointest Liver Physiol 2015; 308:G532-9. [PMID: 25552582 PMCID: PMC4360042 DOI: 10.1152/ajpgi.00281.2014] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Hydrogen sulfide (H₂S) is an endogenous gaseous mediator affecting many physiological and pathophysiological conditions. Enhanced expression of H2S and reactive nitrogen/oxygen species (RNS/ROS) during inflammation alters cellular excitability via modulation of ion channel function. Sulfhydration of cysteine residues and tyrosine nitration are the posttranslational modifications induced by H₂S and RNS, respectively. The objective of this study was to define the interaction between tyrosine nitration and cysteine sulfhydration within the ATP-sensitive K(+) (KATP) channel complex, a significant target in experimental colitis. A modified biotin switch assay was performed to determine sulfhydration of the KATP channel subunits, Kir6.1, sulphonylurea 2B (SUR2B), and nitrotyrosine measured by immunoblot. NaHS (a donor of H₂S) significantly enhanced sulfhydration of SUR2B but not Kir6.1 subunit. 3-Morpholinosydnonimine (SIN-1) (a donor of peroxynitrite) induced nitration of Kir6.1 subunit but not SUR2B. Pretreatment with NaHS reduced the nitration of Kir6.1 by SIN-1 in Chinese hamster ovary cells cotransfected with the two subunits, as well as in enteric glia. Two specific mutations within SUR2B, C24S, and C1455S prevented sulfhydration by NaHS, and these mutations prevented NaHS-induced reduction in tyrosine nitration of Kir6.1. NaHS also reversed peroxynitrite-induced inhibition of smooth muscle contraction. These studies suggest that posttranslational modifications of the two subunits of the KATP channel interact to alter channel function. The studies described herein demonstrate a unique mechanism by which sulfhydration of one subunit modifies tyrosine nitration of another subunit within the same channel complex. This interaction provides a mechanistic insight on the protective effects of H₂S in inflammation.
Collapse
Affiliation(s)
- Minho Kang
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - Atsushi Hashimoto
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - Aravind Gade
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| | - Hamid I. Akbarali
- Department of Pharmacology and Toxicology, Medical College of Virginia Campus, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
36
|
Nalli AD, Rajagopal S, Mahavadi S, Grider JR, Murthy KS. Inhibition of RhoA-dependent pathway and contraction by endogenous hydrogen sulfide in rabbit gastric smooth muscle cells. Am J Physiol Cell Physiol 2015; 308:C485-95. [PMID: 25567809 DOI: 10.1152/ajpcell.00280.2014] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Inhibitory neurotransmitters, chiefly nitric oxide and vasoactive intestinal peptide, increase cyclic nucleotide levels and inhibit muscle contraction via inhibition of myosin light chain (MLC) kinase and activation of MLC phosphatase (MLCP). H2S produced as an endogenous signaling molecule synthesized mainly from l-cysteine via cystathionine-γ-lyase (CSE) and cystathionine-β-synthase (CBS) regulates muscle contraction. The aim of this study was to analyze the expression of CSE and H2S function in the regulation of MLCP activity, 20-kDa regulatory light chain of myosin II (MLC20) phosphorylation, and contraction in isolated gastric smooth muscle cells. Both mRNA expression and protein expression of CSE, but not CBS, were detected in smooth muscle cells of rabbit, human, and mouse stomach. l-cysteine, an activator of CSE, and NaHS, a donor of H2S, inhibited carbachol-induced Rho kinase and PKC activity, Rho kinase-sensitive phosphorylation of MYPT1, PKC-sensitive phosphorylation of CPI-17, and MLC20 phosphorylation and sustained muscle contraction. The inhibitory effects of l-cysteine, but not NaHS, were blocked upon suppression of CSE expression by siRNA or inhibition of its activity by dl-propargylglycine (PPG) suggesting that the effect of l-cysteine is mediated via activation of CSE. Glibenclamide, an inhibitor of KATP channels, had no effect on the inhibition of contraction by H2S. Both l-cysteine and NaHS had no effect on basal cAMP and cGMP levels but augmented forskolin-induced cAMP and SNP-induced cGMP formation. We conclude that both endogenous and exogenous H2S inhibit muscle contraction, and the mechanism involves inhibition of Rho kinase and PKC activities and stimulation of MLCP activity leading to MLC20 dephosphorylation and inhibition of muscle contraction.
Collapse
Affiliation(s)
- Ancy D Nalli
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Senthilkumar Rajagopal
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Sunila Mahavadi
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - John R Grider
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Karnam S Murthy
- Department of Physiology and Biophysics, Virginia Commonwealth University Program in Enteric Neuromuscular Sciences, Virginia Commonwealth University, Richmond, Virginia
| |
Collapse
|
37
|
Pozsgai G, Benkó R, Barthó L, Horváth K, Pintér E. Thermal spring water drinking attenuates dextran-sulfate-sodium-induced colitis in mice. Inflammopharmacology 2015; 23:57-64. [PMID: 25556814 DOI: 10.1007/s10787-014-0227-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 12/22/2014] [Indexed: 01/23/2023]
Abstract
INTRODUCTION The present study investigates the effect of oral consumption of hydrogen sulfide-containing Harkány thermal spring water, as well as sodium hydrogen sulfide (NaHS) solution on experimental colitis. METHODS Colitis was induced by 2% dextran sulfate sodium (DSS) in the drinking water of C57BL/6 mice for 7 days. Some animal groups drank Harkány thermal spring water or water supplemented with 21.68 mg/L NaHS. General signs of colitis, myeloperoxidase (MPO) enzyme activity of colon samples, histological features of colitis and function of the enteric nervous system were assessed. RESULTS Oral administration of Harkány thermal spring water significantly attenuated general signs of colitis, MPO enzyme activity of colon samples and detrimental effect of colitis on the function of the enteric nervous system, but not histological signs of colitis. These findings could be reproduced using NaHS solution with additional significantly diminished histological damage. CONCLUSIONS We conclude that oral treatment with Harkány thermal spring water relieves various aspects of DSS-evoked colitis in mice. This effect is most likely to be mediated by hydrogen sulfide content of the Harkány water. Our data might promote complementary utilization of sulfurous thermal spring water in the therapy of inflammatory bowel disease.
Collapse
Affiliation(s)
- Gábor Pozsgai
- Department of Pharmacology and Pharmacotherapy, University of Pécs, Szigeti u. 12, Pécs, 7624, Hungary,
| | | | | | | | | |
Collapse
|
38
|
Olas B. Hydrogen sulfide in signaling pathways. Clin Chim Acta 2014; 439:212-8. [PMID: 25444740 DOI: 10.1016/j.cca.2014.10.037] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Revised: 10/23/2014] [Accepted: 10/24/2014] [Indexed: 10/24/2022]
Abstract
For a long time hydrogen sulfide (H₂S) was considered a toxic compound, but recently H₂S (at low concentrations) has been found to play an important function in physiological processes. Hydrogen sulfide, like other well-known compounds - nitric oxide (NO) and carbon monoxide (CO) is a gaseous intracellular signal transducer. It regulates the cell cycle, apoptosis and the oxidative stress. Moreover, its functions include neuromodulation, regulation of cardiovascular system and inflammation. In this review, I focus on the metabolism of hydrogen sulfide (including enzymatic pathways of H₂S synthesis from l- and d-cysteine) and its signaling pathways in the cardiovascular system and the nervous system. I also describe how hydrogen sulfide may be used as therapeutic agent, i.e. in the cardiovascular diseases.
Collapse
Affiliation(s)
- Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Lodz, Poland.
| |
Collapse
|
39
|
Farrugia G, Szurszewski JH. Carbon monoxide, hydrogen sulfide, and nitric oxide as signaling molecules in the gastrointestinal tract. Gastroenterology 2014; 147:303-13. [PMID: 24798417 PMCID: PMC4106980 DOI: 10.1053/j.gastro.2014.04.041] [Citation(s) in RCA: 119] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2014] [Revised: 04/16/2014] [Accepted: 04/24/2014] [Indexed: 12/24/2022]
Abstract
Carbon monoxide (CO) and hydrogen sulfide (H2S) used to be thought of simply as lethal and (for H2S) smelly gaseous molecules; now they are known to have important signaling functions in the gastrointestinal tract. CO and H2S, which are produced in the gastrointestinal tract by different enzymes, regulate smooth muscle membrane potential and tone, transmit signals from enteric nerves, and can regulate the immune system. The pathways that produce nitric oxide, H2S, and CO interact; each can inhibit and potentiate the level and activity of the other. However, there are significant differences between these molecules, such as in half-lives; CO is more stable and therefore able to have effects distal to the site of production, whereas nitric oxide and H2S are short lived and act only close to sites of production. We review their signaling functions in the luminal gastrointestinal tract and discuss how their pathways interact. We also describe other physiological functions of CO and H2S and how they might be used as therapeutic agents.
Collapse
Affiliation(s)
- Gianrico Farrugia
- Enteric NeuroScience Program, Division of Gastroenterology and Hepatology and Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, Minnesota.
| | | |
Collapse
|
40
|
Kabil O, Motl N, Banerjee R. H2S and its role in redox signaling. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1844:1355-66. [PMID: 24418393 PMCID: PMC4048824 DOI: 10.1016/j.bbapap.2014.01.002] [Citation(s) in RCA: 163] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Revised: 12/19/2013] [Accepted: 01/02/2014] [Indexed: 02/08/2023]
Abstract
Hydrogen sulfide (H2S) has emerged as an important gaseous signaling molecule that is produced endogenously by enzymes in the sulfur metabolic network. H2S exerts its effects on multiple physiological processes important under both normal and pathological conditions. These functions include neuromodulation, regulation of blood pressure and cardiac function, inflammation, cellular energetics and apoptosis. Despite the recognition of its biological importance and its beneficial effects, the mechanism of H2S action and the regulation of its tissue levels remain unclear in part owing to its chemical and physical properties that render handling and analysis challenging. Furthermore, the multitude of potential H2S effects has made it difficult to dissect its signaling mechanism and to identify specific targets. In this review, we focus on H2S metabolism and provide an overview of the recent literature that sheds some light on its mechanism of action in cellular redox signaling in health and disease. This article is part of a Special Issue entitled: Thiol-Based Redox Processes.
Collapse
Affiliation(s)
- Omer Kabil
- University of Michigan Medical School, Ann Arbor, MI 48109-0600, USA
| | - Nicole Motl
- University of Michigan Medical School, Ann Arbor, MI 48109-0600, USA
| | - Ruma Banerjee
- University of Michigan Medical School, Ann Arbor, MI 48109-0600, USA.
| |
Collapse
|
41
|
Zhao AP, Dong YF, Liu W, Gu J, Sun XL. Nicorandil inhibits inflammasome activation and Toll-like receptor-4 signal transduction to protect against oxygen-glucose deprivation-induced inflammation in BV-2 cells. CNS Neurosci Ther 2013; 20:147-53. [PMID: 24256503 DOI: 10.1111/cns.12178] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2013] [Revised: 08/11/2013] [Accepted: 08/12/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSE Our previous studies have demonstrated adenosine triphosphate-sensitive potassium channel (KATP channel) openers could protect against inflammatory response in brain disease, but little is known about the mechanisms involved in KATP channel openers inhibiting neuroinflammation. METHODS AND RESULTS In the present study, we found that oxygen-glucose deprivation (OGD) resulted in BV-2 cells activation, significantly increased tumor necrosis factor-alpha and interleukin-1beta (IL-1β) levels, accompanied by downregulating Kir6.1 subunit. Pretreatment with nicorandil, a KATP channel opener, could attenuate OGD-induced BV-2 cells activation and inhibit pro-inflammatory factors release. Further study demonstrated that OGD activated Toll-like receptor-4 (TLR4) signaling pathway and NOD-like receptor pyrin domain containing three inflammasome, thereby increased IL-1β production. Pretreatment with nicorandil could reverse the two pathways involved in IL-1β production. CONCLUSIONS Our findings reveal that KATP channel openers could protect against OGD-induced neuroinflammation via inhibiting inflammasome activation and TLR4 signal transduction.
Collapse
Affiliation(s)
- An-Peng Zhao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | | | | | | | | |
Collapse
|
42
|
Low D, Mizoguchi A, Mizoguchi E. DNA methylation in inflammatory bowel disease and beyond. World J Gastroenterol 2013; 19:5238-5249. [PMID: 23983426 PMCID: PMC3752557 DOI: 10.3748/wjg.v19.i32.5238] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2013] [Revised: 05/13/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a consequence of the complex, dysregulated interplay between genetic predisposition, environmental factors, and microbial composition in the intestine. Despite a great advancement in identifying host-susceptibility genes using genome-wide association studies (GWAS), the majority of IBD cases are still underrepresented. The immediate challenge in post-GWAS era is to identify other causative genetic factors of IBD. DNA methylation has received increasing attention for its mechanistical role in IBD pathogenesis. This stable, yet dynamic DNA modification, can directly affect gene expression that have important implications in IBD development. The alterations in DNA methylation associated with IBD are likely to outset as early as embryogenesis all the way until old-age. In this review, we will discuss the recent advancement in understanding how DNA methylation alterations can contribute to the development of IBD.
Collapse
|
43
|
Lu C, Kavalier A, Lukyanov E, Gross SS. S-sulfhydration/desulfhydration and S-nitrosylation/denitrosylation: a common paradigm for gasotransmitter signaling by H2S and NO. Methods 2013; 62:177-81. [PMID: 23811297 DOI: 10.1016/j.ymeth.2013.05.020] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Accepted: 05/28/2013] [Indexed: 12/20/2022] Open
Abstract
Sulfhydryl groups on protein Cys residues undergo an array of oxidative reactions and modifications, giving rise to a virtual redox zip code with physiological and pathophysiological relevance for modulation of protein structure and functions. While over two decades of studies have established NO-dependent S-nitrosylation as ubiquitous and fundamental for the regulation of diverse protein activities, proteomic methods for studying H2S-dependent S-sulfhydration have only recently been described and now suggest that this is also an abundant modification with potential for global physiological importance. Notably, protein S-sulfhydration and S-nitrosylation bear striking similarities in terms of their chemical and biological determinants, as well as reversal of these modifications via group-transfer to glutathione, followed by the removal from glutathione by enzymes that have apparently evolved to selectively catalyze denitrosylation and desulfhydration. Here we review determinants of protein and low-molecular-weight thiol S-sulfhydration/desulfhydration, similarities with S-nitrosylation/denitrosylation, and methods that are being employed to investigate and quantify these gasotransmitter-mediated cell signaling systems.
Collapse
Affiliation(s)
- Changyuan Lu
- Department of Pharmacology, Weill Cornell College of Medicine, 1300 York Avenue, New York, NY, USA
| | | | | | | |
Collapse
|