1
|
Andrés CMC, Pérez de la Lastra JM, Andrés Juan C, Plou FJ, Pérez-Lebeña E. Chemistry of Hydrogen Sulfide-Pathological and Physiological Functions in Mammalian Cells. Cells 2023; 12:2684. [PMID: 38067112 PMCID: PMC10705518 DOI: 10.3390/cells12232684] [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: 09/18/2023] [Revised: 11/02/2023] [Accepted: 11/20/2023] [Indexed: 12/18/2023] Open
Abstract
Hydrogen sulfide (H2S) was recognized as a gaseous signaling molecule, similar to nitric oxide (-NO) and carbon monoxide (CO). The aim of this review is to provide an overview of the formation of hydrogen sulfide (H2S) in the human body. H2S is synthesized by enzymatic processes involving cysteine and several enzymes, including cystathionine-β-synthase (CBS), cystathionine-γ-lyase (CSE), cysteine aminotransferase (CAT), 3-mercaptopyruvate sulfurtransferase (3MST) and D-amino acid oxidase (DAO). The physiological and pathological effects of hydrogen sulfide (H2S) on various systems in the human body have led to extensive research efforts to develop appropriate methods to deliver H2S under conditions that mimic physiological settings and respond to various stimuli. These functions span a wide spectrum, ranging from effects on the endocrine system and cellular lifespan to protection of liver and kidney function. The exact physiological and hazardous thresholds of hydrogen sulfide (H2S) in the human body are currently not well understood and need to be researched in depth. This article provides an overview of the physiological significance of H2S in the human body. It highlights the various sources of H2S production in different situations and examines existing techniques for detecting this gas.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez, 3, 38206 La Laguna, Spain
| | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén, 7, 47011 Valladolid, Spain;
| | - Francisco J. Plou
- Institute of Catalysis and Petrochemistry, CSIC-Spanish Research Council, 28049 Madrid, Spain;
| | | |
Collapse
|
2
|
Hansen AW, Venkatachalam KV. Sulfur-Element containing metabolic pathways in human health and crosstalk with the microbiome. Biochem Biophys Rep 2023; 35:101529. [PMID: 37601447 PMCID: PMC10439400 DOI: 10.1016/j.bbrep.2023.101529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 08/09/2023] [Accepted: 08/10/2023] [Indexed: 08/22/2023] Open
Abstract
In humans, methionine derived from dietary proteins is necessary for cellular homeostasis and regeneration of sulfur containing pathways, which produce inorganic sulfur species (ISS) along with essential organic sulfur compounds (OSC). In recent years, inorganic sulfur species have gained attention as key players in the crosstalk of human health and the gut microbiome. Endogenously, ISS includes hydrogen sulfide (H2S), sulfite (SO32-), thiosulfate (S2O32-), and sulfate (SO42-), which are produced by enzymes in the transsulfuration and sulfur oxidation pathways. Additionally, sulfate-reducing bacteria (SRB) in the gut lumen are notable H2S producers which can contribute to the ISS pools of the human host. In this review, we will focus on the systemic effects of sulfur in biological pathways, describe the contrasting mechanisms of sulfurylation versus phosphorylation on the hydroxyl of serine/threonine and tyrosine residues of proteins in post-translational modifications, and the role of the gut microbiome in human sulfur metabolism.
Collapse
Affiliation(s)
- Austin W. Hansen
- College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, 33328, USA
| | | |
Collapse
|
3
|
Alkaissi H, McFarlane SI. Hyperhomocysteinemia and Accelerated Aging: The Pathogenic Role of Increased Homocysteine in Atherosclerosis, Osteoporosis, and Neurodegeneration. Cureus 2023; 15:e42259. [PMID: 37605676 PMCID: PMC10440097 DOI: 10.7759/cureus.42259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/21/2023] [Indexed: 08/23/2023] Open
Abstract
Cardiovascular diseases and osteoporosis, seemingly unrelated disorders that occur with advanced age, share major pathogenetic mechanisms contributing to accelerated atherosclerosis and bone loss. Hyperhomocysteinemia (hHcy) is among these mechanisms that can cause both vascular and bone disease. In its more severe form, hHcy can present early in life as homocystinuria, an inborn error of metabolic pathways of the sulfur-containing amino acid methionine. In its milder forms, hHcy may go undiagnosed and untreated into adulthood. As such, hHcy may serve as a potential therapeutic target for cardiovascular disease, osteoporosis, thrombophilia, and neurodegeneration, collectively representing accelerated aging. Multiple trials to lower cardiovascular risk and improve bone density with homocysteine-lowering agents, yet none has proven to be clinically meaningful. To understand this unmet clinical need, this review will provide mechanistic insight into the pathogenesis of vascular and bone disease in hHcy, using homocystinuria as a model for accelerated atherosclerosis and bone density loss, a model for accelerated aging.
Collapse
Affiliation(s)
- Hussam Alkaissi
- Internal Medicine, Kings County Hospital Center, Brooklyn, USA
- Internal Medicine, Veterans Affairs Medical Center, Brooklyn, USA
- Internal Medicine, State University of New York Downstate Medical Center, Brooklyn, USA
| | - Samy I McFarlane
- Endocrinology, State University of New York Downstate Medical Center, Brooklyn, USA
| |
Collapse
|
4
|
Jiang J, Wen C, Li Y, Liu G, Chen Z, Zheng D. IFC-305 attenuates renal ischemia-reperfusion injury by promoting the production of hydrogen sulfide (H 2S) via suppressing the promoter methylation of cystathionine γ-lyase (CSE). Bioengineered 2022; 13:12045-12054. [PMID: 35549822 PMCID: PMC9275864 DOI: 10.1080/21655979.2022.2062105] [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] [Indexed: 11/20/2022] Open
Abstract
Renal ischemia-reperfusion (I/R) injury is characterized by elevated expression of homocysteine and decreased production of hydrogen sulfide (H2S). Cystathionine γ-lyase (CSE) is a key factor in the onset of renal I/R injury, while IFC-305 can regulate the expression of CSE via epigenetic modification. Animal and cellular models of I/R were established in this work, followed by H&E staining to evaluate the extent of renal tissue injury under distinct conditions. Several methods, including ELISA, qPCR and Western blot, were used to analyze the levels of creatinine, CSE and H2S in various I/R models. Bisulfite sequencing PCR was used to evaluate the level of DNA methylation. The severity of the renal injury was significantly elevated in I/R rats and alleviated by the IFC-305 treatment. The level of Hcy was increased in the renal tissue and peripheral blood of I/R rats, while the IFC-305 treatment inhibited the expression of homocysteine (Hcy). Mechanistically, the DNA methylation in the CSE promoter was dramatically enhanced in I/R rats and cells, while the IFC-305 treatment reduced the level of DNA methylation in the CSE promoter. Moreover, the IFC-305 increased the concentration of H2S, which was reduced in I/R rats and cells. Finally, I/R rats and cells showed aberrantly high levels of MDA and superoxide, while the IFC-305 treatment reduced the levels of malondialdehyde (MDA) and superoxide. IFC-305, an adenosine derivative, promoted the production of H2S and attenuated renal injury in cellular and animal models of renal I/R by modifying the methylation status of the CSE promoter.
Collapse
Affiliation(s)
- Jie Jiang
- Nephrology Department, Dongguan People's Hospital Affiliated to Southern Medical University, Dongguan, China
| | - Chuling Wen
- Nephrology Department, Dongguan People's Hospital Affiliated to Southern Medical University, Dongguan, China
| | - Yi Li
- Nephrology Department, Dongguan People's Hospital Affiliated to Southern Medical University, Dongguan, China
| | - Guohui Liu
- Nephrology Department, Dongguan People's Hospital Affiliated to Southern Medical University, Dongguan, China
| | - Zijun Chen
- Nephrology Department, Dongguan People's Hospital Affiliated to Southern Medical University, Dongguan, China
| | - Dongwen Zheng
- Nephrology Department, Dongguan People's Hospital Affiliated to Southern Medical University, Dongguan, China
| |
Collapse
|
5
|
Arif HM, Qian Z, Wang R. Signaling Integration of Hydrogen Sulfide and Iron on Cellular Functions. Antioxid Redox Signal 2022; 36:275-293. [PMID: 34498949 DOI: 10.1089/ars.2021.0203] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Significance: Hydrogen sulfide (H2S) is an endogenous signaling molecule, regulating numerous physiological functions from vasorelaxation to neuromodulation. Iron is a well-known bioactive metal ion, being the central component of hemoglobin for oxygen transportation and participating in biomolecule degradation, redox balance, and enzymatic actions. The interplay between H2S and iron metabolisms and functions impacts significantly on the fate and wellness of different types of cells. Recent Advances: Iron level in vivo affects the production of H2S via nonenzymatic reactions. On the contrary, H2S quenches excessive iron inside the cells and regulates the redox status of iron. Critical Issues: Abnormal metabolisms of both iron and H2S are associated with various conditions and diseases such as iron overload, anemia, oxidative stress, and cardiovascular and neurodegenerative diseases. The molecular mechanisms for the interactions between H2S and iron are unsettled yet. Here we review signaling links of the production, metabolism, and their respective and integrative functions of H2S and iron in normalcy and diseases. Future Directions: Physiological and pathophysiological importance of H2S and iron as well as their therapeutic applications should be evaluated jointly, not separately. Future investigation should expand from iron-rich cells and tissues to the others, in which H2S and iron interaction has not received due attention. Antioxid. Redox Signal. 36, 275-293.
Collapse
Affiliation(s)
| | - Zhongming Qian
- Institute of Translational & Precision Medicine, Nantong University, Nantong, China
| | - Rui Wang
- Department of Biology, York University, Toronto, Canada
| |
Collapse
|
6
|
Cui T, Liu W, Yu C, Ren J, Li Y, Shi X, Li Q, Zhang J. Protective Effects of Allicin on Acute Myocardial Infarction in Rats via Hydrogen Sulfide-mediated Regulation of Coronary Arterial Vasomotor Function and Myocardial Calcium Transport. Front Pharmacol 2022; 12:752244. [PMID: 35046802 PMCID: PMC8762278 DOI: 10.3389/fphar.2021.752244] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 11/30/2021] [Indexed: 12/12/2022] Open
Abstract
Acute myocardial infarction (AMI) is a condition with high morbidity and mortality, for which effective treatments are lacking. Allicin has been reported to exert therapeutic effects on AMI, but the underlying mechanisms of its action have not been fully elucidated. To investigate this, a rat model of AMI was generated by ligating the left anterior descending branch of the coronary artery. DL-propargylglycine (PAG), a specific hydrogen sulfide (H2S) synthetase inhibitor, was used to examine the effects of allicin on H2S production. Isolated coronary arteries and cardiomyocytes were assessed for vascular reactivity and cellular Ca2+ transport using a multiwire myography system and a cell-contraction-ion detection system, respectively. Allicin administration improved cardiac function and myocardial pathology, reduced myocardial enzyme levels, and increased H2S and H2S synthetase levels. Allicin administration resulted in concentration-dependent effects on coronary artery dilation, which were mediated by receptor-dependent Ca2+ channels, ATP-sensitive K+ channels, and sarcoplasmic reticulum (SR) Ca2+ release induced by the ryanodine receptor. Allicin administration improved Ca2+ homeostasis in cardiomyocytes by increasing cardiomyocyte contraction, Ca2+ transient amplitude, myofilament sensitivity, and SR Ca2+ content. Allicin also enhanced Ca2+ uptake via SR Ca2+-ATPase and Ca2+ removal via the Na+/Ca2+ exchanger, and it reduced SR Ca2+ leakage. Notably, the protective effects of allicin were partially attenuated by blockade of H2S production with PAG. Our findings provide novel evidence that allicin-induced production of H2S mediates coronary artery dilation and regulation of Ca2+ homeostasis in AMI. Our study presents a novel mechanistic insight into the anti-AMI effects of allicin and highlights the therapeutic potential of this compound.
Collapse
Affiliation(s)
- Tianwei Cui
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China.,Department of Reproductive Medicine, The First Affiliated Hospital of Henan University of Chinese Medicine, Zhengzhou, China
| | - Weiyu Liu
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Chenghao Yu
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jianxun Ren
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yikui Li
- Health Prevention Department, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaolu Shi
- Beijing Key Laboratory of TCM Basic Research on Prevention and Treatment of Major Disease, Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qiuyan Li
- Department of General Medicine, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Jinyan Zhang
- Institute of Basic Medical Sciences, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| |
Collapse
|
7
|
Olson KR. A Case for Hydrogen Sulfide Metabolism as an Oxygen Sensing Mechanism. Antioxidants (Basel) 2021; 10:antiox10111650. [PMID: 34829521 PMCID: PMC8615108 DOI: 10.3390/antiox10111650] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 10/08/2021] [Accepted: 10/13/2021] [Indexed: 12/30/2022] Open
Abstract
The ability to detect oxygen availability is a ubiquitous attribute of aerobic organisms. However, the mechanism(s) that transduce oxygen concentration or availability into appropriate physiological responses is less clear and often controversial. This review will make the case for oxygen-dependent metabolism of hydrogen sulfide (H2S) and polysulfides, collectively referred to as reactive sulfur species (RSS) as a physiologically relevant O2 sensing mechanism. This hypothesis is based on observations that H2S and RSS metabolism is inversely correlated with O2 tension, exogenous H2S elicits physiological responses identical to those produced by hypoxia, factors that affect H2S production or catabolism also affect tissue responses to hypoxia, and that RSS efficiently regulate downstream effectors of the hypoxic response in a manner consistent with a decrease in O2. H2S-mediated O2 sensing is then compared to the more generally accepted reactive oxygen species (ROS) mediated O2 sensing mechanism and a number of reasons are offered to resolve some of the confusion between the two.
Collapse
Affiliation(s)
- Kenneth R Olson
- Department of Physiology, Indiana University School of Medicine-South Bend, South Bend, IN 46617, USA
| |
Collapse
|
8
|
Abstract
Hibernation is a powerful response of a number of mammalian species to reduce energy during the cold winter season, when food is scarce. Mammalian hibernators survive winter by spending most of the time in a state of torpor, where basal metabolic rate is strongly suppressed and body temperature comes closer to ambient temperature. These torpor bouts are regularly interrupted by short arousals, where metabolic rate and body temperature spontaneously return to normal levels. The mechanisms underlying these changes, and in particular the strong metabolic suppression of torpor, have long remained elusive. As summarized in this Commentary, increasing evidence points to a potential key role for hydrogen sulfide (H2S) in the suppression of mitochondrial respiration during torpor. The idea that H2S could be involved in hibernation originated in some early studies, where exogenous H2S gas was found to induce a torpor-like state in mice, and despite some controversy, the idea persisted. H2S is a widespread signaling molecule capable of inhibiting mitochondrial respiration in vitro and studies found significant in vivo changes in endogenous H2S metabolites associated with hibernation or torpor. Along with increased expression of H2S-synthesizing enzymes during torpor, H2S degradation catalyzed by the mitochondrial sulfide:quinone oxidoreductase (SQR) appears to have a key role in controlling H2S availability for inhibiting respiration. Specifically, in thirteen-lined squirrels, SQR is highly expressed and inhibited in torpor, possibly by acetylation, thereby limiting H2S oxidation and causing inhibition of respiration. H2S may also control other aspects associated with hibernation, such as synthesis of antioxidant enzymes and of SQR itself.
Collapse
Affiliation(s)
| | - Angela Fago
- Department of Biology, Aarhus University, Aarhus C 8000, Denmark
| |
Collapse
|
9
|
Kuschman HP, Palczewski MB, Thomas DD. Nitric oxide and hydrogen sulfide: Sibling rivalry in the family of epigenetic regulators. Free Radic Biol Med 2021; 170:34-43. [PMID: 33482335 DOI: 10.1016/j.freeradbiomed.2021.01.010] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 12/16/2020] [Accepted: 01/06/2021] [Indexed: 01/12/2023]
Abstract
Nitric oxide (NO) and hydrogen sulfide (H2S) were previously only known for their toxic properties. Now they are regarded as potent gaseous messenger molecules (gasotransmitters) that rapidly transverse cell membranes and transduce cellular signals through their chemical reactions and modifications to protein targets. Both are known to regulate numerous physiological functions including angiogenesis, vascular tone, and immune response, to name a few. NO and H2S often work synergistically and in competition to regulate each other's synthesis, target protein activity via posttranslational modifications (PTMs), and chemical interactions. In addition to their canonical modes of action, increasing evidence has demonstrated that NO and H2S share another signaling mechanism: epigenetic regulation. This review will compare and contrast biosynthesis and metabolism of NO and H2S, their individual and shared interactions, and the growing body of evidence for their roles as endogenous epigenetic regulatory molecules.
Collapse
Affiliation(s)
- Hannah Petraitis Kuschman
- University of Illinois at Chicago, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, United States
| | - Marianne B Palczewski
- University of Illinois at Chicago, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, United States
| | - Douglas D Thomas
- University of Illinois at Chicago, Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL, 60612, United States.
| |
Collapse
|
10
|
Jensen BS, Pardue S, Duffy B, Kevil CG, Staples JF, Fago A. Suppression of mitochondrial respiration by hydrogen sulfide in hibernating 13-lined ground squirrels. Free Radic Biol Med 2021; 169:181-186. [PMID: 33887435 PMCID: PMC8809085 DOI: 10.1016/j.freeradbiomed.2021.04.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/18/2021] [Accepted: 04/08/2021] [Indexed: 12/30/2022]
Abstract
Hibernating mammals may suppress their basal metabolic rate during torpor by up to 95% to reduce energy expenditure during winter, but the underlying mechanisms remain poorly understood. Here we show that hydrogen sulfide (H2S), a ubiquitous signaling molecule, is a powerful inhibitor of respiration of liver mitochondria isolated from torpid 13-lined ground squirrels, but has a weak effect on mitochondria isolated during summer and hibernation arousals, where metabolic rate is normal. Consistent with these in vitro effects, we find strong seasonal variations of in vivo levels of H2S in plasma and increases of H2S levels in the liver of squirrels during torpor compared to levels during arousal and summer. The in vivo changes of liver H2S levels correspond with low activity of the mitochondrial H2S oxidizing enzyme sulfide:quinone oxidoreductase (SQR) during torpor. Taken together, these results suggest that during torpor, H2S accumulates in the liver due to a low SQR activity and contributes to inhibition of mitochondrial respiration, while during arousals and summer these effects are reversed, H2S is degraded by active SQR and mitochondrial respiration rates increase. This study provides novel insights into mechanisms underlying mammalian hibernation, pointing to SQR as a key enzyme involved in the control of mitochondrial function.
Collapse
Affiliation(s)
- Birgitte S Jensen
- Department of Biology, Aarhus University, Aarhus C, 8000, Denmark; Department of Biology, University of Western Ontario, London, ON N6A 5B8, Canada
| | - Sibile Pardue
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - Brynne Duffy
- Department of Biology, University of Western Ontario, London, ON N6A 5B8, Canada
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA, 71130, USA
| | - James F Staples
- Department of Biology, University of Western Ontario, London, ON N6A 5B8, Canada
| | - Angela Fago
- Department of Biology, Aarhus University, Aarhus C, 8000, Denmark.
| |
Collapse
|
11
|
Jiang S, Xu W, Chen Z, Cui C, Fan X, Cai J, Gong Y, Geng B. Hydrogen sulphide reduces hyperhomocysteinaemia-induced endothelial ER stress by sulfhydrating protein disulphide isomerase to attenuate atherosclerosis. J Cell Mol Med 2021; 25:3437-3448. [PMID: 33675119 PMCID: PMC8034471 DOI: 10.1111/jcmm.16423] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/13/2022] Open
Abstract
Hyperhomocysteinaemia (HHcy)-impaired endothelial dysfunction including endoplasmic reticulum (ER) stress plays a crucial role in atherogenesis. Hydrogen sulphide (H2 S), a metabolic production of Hcy and gasotransmitter, exhibits preventing cardiovascular damages induced by HHcy by reducing ER stress, but the underlying mechanism is unclear. Here, we made an atherosclerosis with HHcy mice model by ApoE knockout mice and feeding Pagien diet and drinking L-methionine water. H2 S donors NaHS and GYY4137 treatment lowered plaque area and ER stress in this model. Protein disulphide isomerase (PDI), a modulation protein folding key enzyme, was up-regulated in plaque and reduced by H2 S treatment. In cultured human aortic endothelial cells, Hcy dose and time dependently elevated PDI expression, but inhibited its activity, and which were rescued by H2 S. H2 S and its endogenous generation key enzyme-cystathionine γ lyase induced a new post-translational modification-sulfhydration of PDI. Sulfhydrated PDI enhanced its activity, and two cysteine-terminal CXXC domain of PDI was identified by site mutation. HHcy lowered PDI sulfhydration association ER stress, and H2 S rescued it but this effect was blocked by cysteine site mutation. Conclusively, we demonstrated that H2 S sulfhydrated PDI and enhanced its activity, reducing HHcy-induced endothelial ER stress to attenuate atherosclerosis development.
Collapse
Affiliation(s)
- Shan Jiang
- Institute of Hypoxia Medicine, Wenzhou Medical University, Zhejiang, China
| | - Wenjing Xu
- Department of Pathology, Xi'an Medical University, Shanxi, China
| | - Zhenzhen Chen
- State Key Laboratory of Cardiovascular Disease, Hypertension Center, National Center for Cardiovascular Diseases, Fuwai Hospital of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changting Cui
- State Key Laboratory of Cardiovascular Disease, Hypertension Center, National Center for Cardiovascular Diseases, Fuwai Hospital of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaofang Fan
- Institute of Hypoxia Medicine, Wenzhou Medical University, Zhejiang, China
| | - Jun Cai
- State Key Laboratory of Cardiovascular Disease, Hypertension Center, National Center for Cardiovascular Diseases, Fuwai Hospital of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongsheng Gong
- Institute of Hypoxia Medicine, Wenzhou Medical University, Zhejiang, China
| | - Bin Geng
- Institute of Hypoxia Medicine, Wenzhou Medical University, Zhejiang, China.,State Key Laboratory of Cardiovascular Disease, Hypertension Center, National Center for Cardiovascular Diseases, Fuwai Hospital of Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
12
|
Bonifácio VDB, Pereira SA, Serpa J, Vicente JB. Cysteine metabolic circuitries: druggable targets in cancer. Br J Cancer 2021; 124:862-879. [PMID: 33223534 PMCID: PMC7921671 DOI: 10.1038/s41416-020-01156-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 31.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 09/03/2020] [Accepted: 10/22/2020] [Indexed: 02/07/2023] Open
Abstract
To enable survival in adverse conditions, cancer cells undergo global metabolic adaptations. The amino acid cysteine actively contributes to cancer metabolic remodelling on three different levels: first, in its free form, in redox control, as a component of the antioxidant glutathione or its involvement in protein s-cysteinylation, a reversible post-translational modification; second, as a substrate for the production of hydrogen sulphide (H2S), which feeds the mitochondrial electron transfer chain and mediates per-sulphidation of ATPase and glycolytic enzymes, thereby stimulating cellular bioenergetics; and, finally, as a carbon source for epigenetic regulation, biomass production and energy production. This review will provide a systematic portrayal of the role of cysteine in cancer biology as a source of carbon and sulphur atoms, the pivotal role of cysteine in different metabolic pathways and the importance of H2S as an energetic substrate and signalling molecule. The different pools of cysteine in the cell and within the body, and their putative use as prognostic cancer markers will be also addressed. Finally, we will discuss the pharmacological means and potential of targeting cysteine metabolism for the treatment of cancer.
Collapse
Affiliation(s)
- Vasco D B Bonifácio
- iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Avenida Rovisco Pais, 1049-001, Lisboa, Portugal
| | - Sofia A Pereira
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal
| | - Jacinta Serpa
- CEDOC, Chronic Diseases Research Centre, NOVA Medical School Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Campo dos Mártires da Pátria, 130, 1169-056, Lisboa, Portugal.
- Instituto Português de Oncologia de Lisboa Francisco Gentil (IPOLFG), Rua Prof Lima Basto, 1099-023, Lisboa, Portugal.
| | - João B Vicente
- Instituto de Tecnologia Química e Biológica António Xavier (ITQB NOVA), Avenida da República (EAN), 2780-157, Oeiras, Portugal
| |
Collapse
|
13
|
Giroud S, Habold C, Nespolo RF, Mejías C, Terrien J, Logan SM, Henning RH, Storey KB. The Torpid State: Recent Advances in Metabolic Adaptations and Protective Mechanisms †. Front Physiol 2021; 11:623665. [PMID: 33551846 PMCID: PMC7854925 DOI: 10.3389/fphys.2020.623665] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Accepted: 12/21/2020] [Indexed: 12/18/2022] Open
Abstract
Torpor and hibernation are powerful strategies enabling animals to survive periods of low resource availability. The state of torpor results from an active and drastic reduction of an individual's metabolic rate (MR) associated with a relatively pronounced decrease in body temperature. To date, several forms of torpor have been described in all three mammalian subclasses, i.e., monotremes, marsupials, and placentals, as well as in a few avian orders. This review highlights some of the characteristics, from the whole organism down to cellular and molecular aspects, associated with the torpor phenotype. The first part of this review focuses on the specific metabolic adaptations of torpor, as it is used by many species from temperate zones. This notably includes the endocrine changes involved in fat- and food-storing hibernating species, explaining biomedical implications of MR depression. We further compare adaptive mechanisms occurring in opportunistic vs. seasonal heterotherms, such as tropical and sub-tropical species. Such comparisons bring new insights into the metabolic origins of hibernation among tropical species, including resistance mechanisms to oxidative stress. The second section of this review emphasizes the mechanisms enabling heterotherms to protect their key organs against potential threats, such as reactive oxygen species, associated with the torpid state. We notably address the mechanisms of cellular rehabilitation and protection during torpor and hibernation, with an emphasis on the brain, a central organ requiring protection during torpor and recovery. Also, a special focus is given to the role of an ubiquitous and readily-diffusing molecule, hydrogen sulfide (H2S), in protecting against ischemia-reperfusion damage in various organs over the torpor-arousal cycle and during the torpid state. We conclude that (i) the flexibility of torpor use as an adaptive strategy enables different heterothermic species to substantially suppress their energy needs during periods of severely reduced food availability, (ii) the torpor phenotype implies marked metabolic adaptations from the whole organism down to cellular and molecular levels, and (iii) the torpid state is associated with highly efficient rehabilitation and protective mechanisms ensuring the continuity of proper bodily functions. Comparison of mechanisms in monotremes and marsupials is warranted for understanding the origin and evolution of mammalian torpor.
Collapse
Affiliation(s)
- Sylvain Giroud
- Research Institute of Wildlife Ecology, Department of Interdisciplinary Life Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Caroline Habold
- University of Strasbourg, CNRS, IPHC, UMR 7178, Strasbourg, France
| | - Roberto F. Nespolo
- Instituto de Ciencias Ambientales y Evolutivas, Universidad Austral de Chile, ANID – Millennium Science Initiative Program-iBio, Valdivia, Chile
- Center of Applied Ecology and Sustainability, Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Carlos Mejías
- Instituto de Ciencias Ambientales y Evolutivas, Universidad Austral de Chile, ANID – Millennium Science Initiative Program-iBio, Valdivia, Chile
- Center of Applied Ecology and Sustainability, Departamento de Ecología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jérémy Terrien
- Unité Mécanismes Adaptatifs et Evolution (MECADEV), UMR 7179, CNRS, Muséum National d’Histoire Naturelle, Brunoy, France
| | | | - Robert H. Henning
- Department of Clinical Pharmacy and Pharmacology, University Medical Center Groningen, Groningen, Netherlands
| | | |
Collapse
|
14
|
Transsulfuration metabolites and the association with incident atrial fibrillation – An observational cohort study among Norwegian patients with stable angina pectoris. Int J Cardiol 2020; 317:75-80. [DOI: 10.1016/j.ijcard.2020.05.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/16/2020] [Accepted: 05/04/2020] [Indexed: 11/19/2022]
|
15
|
Liu N, Lin X, Huang C. Activation of the reverse transsulfuration pathway through NRF2/CBS confers erastin-induced ferroptosis resistance. Br J Cancer 2019; 122:279-292. [PMID: 31819185 PMCID: PMC7052275 DOI: 10.1038/s41416-019-0660-x] [Citation(s) in RCA: 150] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/02/2019] [Accepted: 11/08/2019] [Indexed: 12/26/2022] Open
Abstract
Background Ferroptosis is an iron-dependent, lipid peroxide-mediated cell death that may be exploited to selective elimination of damaged and malignant cells. Recent studies have identified that small-molecule erastin specifically inhibits transmembrane cystine–glutamate antiporter system xc−, prevents extracellular cystine import and ultimately causes ferroptosis in certain cancer cells. In this study, we aimed to investigate the molecular mechanism underlying erastin-induced ferroptosis resistance in ovarian cancer cells. Methods We treated ovarian cancer cells with erastin and examined cell viability, cellular ROS and metabolites of the transsulfuration pathway. We also depleted cystathionine β-synthase (CBS) and NRF2 to investigate the CBS and NRF2 dependency in erastin-resistant cells. Results We found that prolonged erastin treatment induced ferroptosis resistance. Upon exposure to erastin, cells gradually adapted to cystine deprivation via sustained activation of the reverse transsulfuration pathway, allowing the cells to bypass erastin insult. CBS, the biosynthetic enzyme for cysteine, was constantly upregulated and was critical for the resistance. Knockdown of CBS by RNAi in erastin-resistant cells caused ferroptotic cell death, while CBS overexpression conferred ferroptosis resistance. We determined that the antioxidant transcriptional factor, NRF2 was constitutively activated in erastin-resistant cells and NRF2 transcriptionally upregulated CBS. Genetically repression of NRF2 enhanced ferroptosis susceptibility. Conclusions Based on these results, we concluded that constitutive activation of NRF2/CBS signalling confers erastin-induced ferroptosis resistance. This study demonstrates a new mechanism underlying ferroptosis resistance, and has implications for the therapeutic response to erastin-induced ferroptosis.
Collapse
Affiliation(s)
- Nan Liu
- Division of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Xiaoli Lin
- Division of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chengying Huang
- Division of Obstetrics and Gynecology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
16
|
Murphy B, Bhattacharya R, Mukherjee P. Hydrogen sulfide signaling in mitochondria and disease. FASEB J 2019; 33:13098-13125. [PMID: 31648556 PMCID: PMC6894098 DOI: 10.1096/fj.201901304r] [Citation(s) in RCA: 163] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2019] [Accepted: 09/17/2019] [Indexed: 02/06/2023]
Abstract
Hydrogen sulfide can signal through 3 distinct mechanisms: 1) reduction and/or direct binding of metalloprotein heme centers, 2) serving as a potent antioxidant through reactive oxygen species/reactive nitrogen species scavenging, or 3) post-translational modification of proteins by addition of a thiol (-SH) group onto reactive cysteine residues: a process known as persulfidation. Below toxic levels, hydrogen sulfide promotes mitochondrial biogenesis and function, thereby conferring protection against cellular stress. For these reasons, increases in hydrogen sulfide and hydrogen sulfide-producing enzymes have been implicated in several human disease states. This review will first summarize our current understanding of hydrogen sulfide production and metabolism, as well as its signaling mechanisms; second, this work will detail the known mechanisms of hydrogen sulfide in the mitochondria and the implications of its mitochondrial-specific impacts in several pathologic conditions.-Murphy, B., Bhattacharya, R., Mukherjee, P. Hydrogen sulfide signaling in mitochondria and disease.
Collapse
Affiliation(s)
- Brennah Murphy
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Resham Bhattacharya
- Department of Obstetrics and Gynecology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Priyabrata Mukherjee
- Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
- Peggy and Charles Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
17
|
Zaorska E, Hutsch T, Gawryś-Kopczyńska M, Ostaszewski R, Ufnal M, Koszelewski D. Evaluation of thioamides, thiolactams and thioureas as hydrogen sulfide (H2S) donors for lowering blood pressure. Bioorg Chem 2019; 88:102941. [DOI: 10.1016/j.bioorg.2019.102941] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/08/2019] [Accepted: 04/18/2019] [Indexed: 01/15/2023]
|
18
|
Rajendran S, Shen X, Glawe J, Kolluru GK, Kevil CG. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Growth and Remodeling. Compr Physiol 2019; 9:1213-1247. [PMID: 31187898 DOI: 10.1002/cphy.c180026] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Ischemic vascular remodeling occurs in response to stenosis or arterial occlusion leading to a change in blood flow and tissue perfusion. Altered blood flow elicits a cascade of molecular and cellular physiological responses leading to vascular remodeling of the macro- and micro-circulation. Although cellular mechanisms of vascular remodeling such as arteriogenesis and angiogenesis have been studied, therapeutic approaches in these areas have had limited success due to the complexity and heterogeneous constellation of molecular signaling events regulating these processes. Understanding central molecular players of vascular remodeling should lead to a deeper understanding of this response and aid in the development of novel therapeutic strategies. Hydrogen sulfide (H2 S) and nitric oxide (NO) are gaseous signaling molecules that are critically involved in regulating fundamental biochemical and molecular responses necessary for vascular growth and remodeling. This review examines how NO and H2 S regulate pathophysiological mechanisms of angiogenesis and arteriogenesis, along with important chemical and experimental considerations revealed thus far. The importance of NO and H2 S bioavailability, their synthesis enzymes and cofactors, and genetic variations associated with cardiovascular risk factors suggest that they serve as pivotal regulators of vascular remodeling responses. © 2019 American Physiological Society. Compr Physiol 9:1213-1247, 2019.
Collapse
Affiliation(s)
| | - Xinggui Shen
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - John Glawe
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - Gopi K Kolluru
- Departments of Pathology, LSU Health Sciences Center, Shreveport
| | - Christopher G Kevil
- Departments of Pathology, LSU Health Sciences Center, Shreveport.,Departments of Cellular Biology and Anatomy, LSU Health Sciences Center, Shreveport.,Departments of Molecular and Cellular Physiology, LSU Health Sciences Center, Shreveport
| |
Collapse
|
19
|
Yang J, Minkler P, Grove D, Wang R, Willard B, Dweik R, Hine C. Non-enzymatic hydrogen sulfide production from cysteine in blood is catalyzed by iron and vitamin B 6. Commun Biol 2019; 2:194. [PMID: 31123718 PMCID: PMC6529520 DOI: 10.1038/s42003-019-0431-5] [Citation(s) in RCA: 124] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 04/18/2019] [Indexed: 12/12/2022] Open
Abstract
Hydrogen sulfide (H2S) plays important roles in metabolism and health. Its enzymatic generation from sulfur-containing amino acids (SAAs) is well characterized. However, the existence of non-enzymatic H2S production from SAAs, the chemical mechanism, and its biological implications remain unclear. Here we present non-enzymatic H2S production in vitro and in blood via a reaction specific for the SAA cysteine serving as substrate and requires coordinated catalysis by Vitamin B6, pyridoxal(phosphate), and iron under physiological conditions. An initial cysteine-aldimine is formed by nucleophilic attack of the cysteine amino group to the pyridoxal(phosphate) aldehyde group. Free or heme-bound iron drives the formation of a cysteine-quinonoid, thiol group elimination, and hydrolysis of the desulfurated aldimine back to pyridoxal(phosphate). The reaction ultimately produces pyruvate, NH3, and H2S. This work highlights enzymatic production is inducible and robust in select tissues, whereas iron-catalyzed production contributes underappreciated basal H2S systemically with pathophysiological implications in hemolytic, iron overload, and hemorrhagic disorders.
Collapse
Affiliation(s)
- Jie Yang
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195 USA
| | - Paul Minkler
- Proteomics and Metabolomics Core, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195 USA
| | - David Grove
- Department of Inflammation and Immunity, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195 USA
| | - Rui Wang
- Faculty of Science, Department of Biology, York University, Toronto, Canada M3J 1P3
| | - Belinda Willard
- Proteomics and Metabolomics Core, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195 USA
| | - Raed Dweik
- Respiratory Institute, Cleveland Clinic, Cleveland, OH 44195 USA
| | - Christopher Hine
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195 USA
| |
Collapse
|
20
|
Badiei A, Sudharsan R, Santana E, Dunaief JL, Aguirre GD. Comparative localization of cystathionine beta synthases and cystathionine gamma lyase in canine, non-human primate and human retina. Exp Eye Res 2019; 181:72-84. [PMID: 30653965 PMCID: PMC6443508 DOI: 10.1016/j.exer.2019.01.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2018] [Revised: 12/18/2018] [Accepted: 01/03/2019] [Indexed: 12/15/2022]
Abstract
Chronic exposure of the retina to light and high concentrations of polyunsaturated fatty acid in photoreceptor cells make this tissue susceptible to oxidative damage. As retinal degenerative diseases are associated with photoreceptor degeneration, the antioxidant activity of both hydrogen sulfide (H2S) and glutathione (GSH) may play an important role in ameliorating disease progression. H2S production is driven by cystathionine-γ-lyase (CSE) and cystathionine-β-synthase (CBS), the key enzymes that also drive transsulfuration pathway (TSP) necessary for GSH production. As it is currently unclear whether localized production of either H2S or GSH contributes to retinal homeostasis, we undertook a comparative analysis of CBS and CSE expression in canine, non-human primate (NHP) and human retinas to determine if these antioxidants could play a regulatory role in age-related or disease-associated retinal degeneration. Retinas from normal dogs, NHPs and humans were used for the study. Laser capture microdissection (LCM) was performed to isolate individual layers of the canine retina and analyze CBS and CSE gene expression by qRT-PCR. Immunohistochemistry and western blotting were performed for CBS and CSE labeling and protein expression in dog, NHP, and human retina, respectively. Using qRT-PCR, western blot, and immunohistochemistry (IHC), we showed that CBS and CSE are expressed in the canine, NHP, and human retina. IHC results from canine retina demonstrated increased expression levels of CBS but not CSE with post-developmental aging. IHC results also showed non-overlapping localization of both proteins with CBS presenting in rods, amacrine, horizontal, and nerve fiber cell layers while CSE was expressed by RPE, cones and Mϋller cells. Finally, we demonstrated that these enzymes localized to all three layers of canine, NHP and human retina: photoreceptors, outer plexiform layer (OPL) and notably in the ganglion cells layer/nerve fiber layer (GCL/NFL). QRT-PCR performed using RNA extracted from tissues isolated from these cell layers using laser capture microdissection (LCM) confirmed that each of CBS and CSE are expressed equally in these three layers. Together, these findings reveal that CSE and CBS are expressed in the retina, thereby supporting further studies to determine the role of H2S and these proteins in oxidative stress and apoptosis in retinal degenerative diseases.
Collapse
Affiliation(s)
- Alireza Badiei
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Raghavi Sudharsan
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Evelyn Santana
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Joshua L Dunaief
- F. M. Kirby Center for Molecular Ophthalmology, Scheie Eye Institute, University of Pennsylvania, Philadelphia, PA, USA
| | - Gustavo D Aguirre
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
21
|
de León Bautista MP, Romero-Valdovinos M, Zavaleta-Villa B, Martínez-Flores A, Olivo-Díaz A. Association of Cystathionine β-Synthase Gene Polymorphisms With Preeclampsia. Clin Appl Thromb Hemost 2018; 24:285S-293S. [PMID: 30380942 PMCID: PMC6714820 DOI: 10.1177/1076029618808913] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Preeclampsia (PE) is a pregnancy disorder that increases maternal and fetal
morbidity and mortality worldwide. High plasma levels of homocysteine (Hcy) are
a risk factor for several cardiovascular diseases. Cystathionine β-synthase
(CBS) plays an important role in Hcy homeostasis catalyzing the irreversible
degradation of Hcy to cystathionine, protecting the endothelium from injury
caused by hypoxia. Several mutations and polymorphisms may alter the expression
of the CBS gene, resulting in variable levels of Hcy. The
purpose of this study was to investigate the association of CBS
gene polymorphisms with PE in Mexican women. A case–control study consisting of
129 pregnant women with PE (37 severe and 92 mild) and 173 women with
uncomplicated pregnancies was performed. Polymorphisms, such as G797A, C785T,
T833C, G919A, T959C, C1105T, and 844ins68 base pair, in the CBS
gene were genotyped. The polymorphism G797A was monomorphic in cases with the
presence of only G797A-G allele. Allele C785T-T and genotype C785T-C/T were
associated with susceptibility in severe and mild PE. Alleles G797A-G and
T959C-T were associated with susceptibility only in severe PE. Haplotype TGTWGTC
was of susceptibility for severe PE and of protection for mild PE. Haplotypes
CGTWGCC and CATWGTC seem to be protective for severe PE, but the latter is
related to susceptibility in mild PE. The results suggest that C785T, G797A, and
T959C mutations are contributing in different ways in severe and mild PE in our
population and could be count as another related factor for this disease.
Collapse
Affiliation(s)
| | - Mirza Romero-Valdovinos
- Department of Molecular Biology and Histocompatibility, General Hospital "Dr Manuel Gea Gonzalez," Mexico City, Mexico
| | - Beatriz Zavaleta-Villa
- Department of Molecular Biology and Histocompatibility, General Hospital "Dr Manuel Gea Gonzalez," Mexico City, Mexico
| | - Arony Martínez-Flores
- Department of Ecology of Pathogens Agents, General Hospital "Dr Manuel Gea Gonzalez," Mexico City, Mexico
| | - Angélica Olivo-Díaz
- Department of Molecular Biology and Histocompatibility, General Hospital "Dr Manuel Gea Gonzalez," Mexico City, Mexico
| |
Collapse
|
22
|
Corsello T, Komaravelli N, Casola A. Role of Hydrogen Sulfide in NRF2- and Sirtuin-Dependent Maintenance of Cellular Redox Balance. Antioxidants (Basel) 2018; 7:antiox7100129. [PMID: 30274149 PMCID: PMC6210431 DOI: 10.3390/antiox7100129] [Citation(s) in RCA: 107] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 09/21/2018] [Accepted: 09/27/2018] [Indexed: 12/18/2022] Open
Abstract
Hydrogen sulfide (H2S) has arisen as a critical gasotransmitter signaling molecule modulating cellular biological events related to health and diseases in heart, brain, liver, vascular systems and immune response. Three enzymes mediate the endogenous production of H2S: cystathione β-synthase (CBS), cystathione γ-lyase (CSE) and 3-mercaptopyruvate sulfurtransferase (3-MST). CBS and CSE localizations are organ-specific. 3-MST is a mitochondrial and cytosolic enzyme. The generation of H2S is firmly regulated by these enzymes under normal physiological conditions. Recent studies have highlighted the role of H2S in cellular redox homeostasis, as it displays significant antioxidant properties. H2S exerts antioxidant effects through several mechanisms, such as quenching reactive oxygen species (ROS) and reactive nitrogen species (RNS), by modulating cellular levels of glutathione (GSH) and thioredoxin (Trx-1) or increasing expression of antioxidant enzymes (AOE), by activating the transcription factor nuclear factor (erythroid-derived 2)-like 2 (NRF2). H2S also influences the activity of the histone deacetylase protein family of sirtuins, which plays an important role in inhibiting oxidative stress in cardiomyocytes and during the aging process by modulating AOE gene expression. This review focuses on the role of H2S in NRF2 and sirtuin signaling pathways as they are related to cellular redox homeostasis.
Collapse
Affiliation(s)
- Tiziana Corsello
- Department of Pediatrics, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| | - Narayana Komaravelli
- Department of Pediatrics, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| | - Antonella Casola
- Department of Pediatrics, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| |
Collapse
|
23
|
Cao X, Xiong S, Zhou Y, Wu Z, Ding L, Zhu Y, Wood ME, Whiteman M, Moore PK, Bian JS. Renal Protective Effect of Hydrogen Sulfide in Cisplatin-Induced Nephrotoxicity. Antioxid Redox Signal 2018; 29:455-470. [PMID: 29316804 DOI: 10.1089/ars.2017.7157] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
AIMS Cisplatin is a major therapeutic drug for solid tumors, but can cause severe nephrotoxicity. However, the role and therapeutic potential of hydrogen sulfide (H2S), an endogenous gasotransmitter, in cisplatin-induced nephrotoxicity remain to be defined. RESULTS Cisplatin led to the impairment of H2S production in vitro and in vivo by downregulating the expression level of cystathionine γ-lyase (CSE), which may contribute to the subsequent renal proximal tubule (RPT) cell death and thereby renal toxicity. H2S donors NaHS and GYY4137, but not AP39, mitigated cisplatin-induced RPT cell death and nephrotoxicity. The mechanisms underlying the protective effect of H2S donors included the suppression of intracellular reactive oxygen species generation and downstream mitogen-activated protein kinases by inhibiting NADPH oxidase activity, which may be possibly through persulfidating the subunit p47phox. Importantly, GYY4137 not only ameliorated cisplatin-caused renal injury but also added on more anticancer effect to cisplatin in cancer cell lines. Innovation and Conclusion: Our study provides a comprehensive understanding of the role and therapeutic potential of H2S in cisplatin-induced nephrotoxicity. Our results indicate that H2S may be a novel and promising therapeutic target to prevent cisplatin-induced nephrotoxicity. Antioxid. Redox Signal. 29, 455-470.
Collapse
Affiliation(s)
- Xu Cao
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Siping Xiong
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Yebo Zhou
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Zhiyuan Wu
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,2 Life Science Institute, National University of Singapore , Singapore, Singapore
| | - Lei Ding
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Yike Zhu
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore
| | - Mark E Wood
- 3 Department of Biosciences, University of Exeter , Exeter, United Kingdom
| | - Matthew Whiteman
- 4 School of Biosciences, College of Life and Environmental Science, University of Exeter , Exeter, United Kingdom
| | - Philip K Moore
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,2 Life Science Institute, National University of Singapore , Singapore, Singapore
| | - Jin-Song Bian
- 1 Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore , Singapore, Singapore .,2 Life Science Institute, National University of Singapore , Singapore, Singapore
| |
Collapse
|
24
|
Implications of plasma thiol redox in disease. Clin Sci (Lond) 2018; 132:1257-1280. [DOI: 10.1042/cs20180157] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 05/09/2018] [Accepted: 05/22/2018] [Indexed: 12/21/2022]
Abstract
Thiol groups are crucially involved in signaling/homeostasis through oxidation, reduction, and disulphide exchange. The overall thiol pool is the resultant of several individual pools of small compounds (e.g. cysteine), peptides (e.g. glutathione), and thiol proteins (e.g. thioredoxin (Trx)), which are not in equilibrium and present specific oxidized/reduced ratios. This review addresses mechanisms and implications of circulating plasma thiol/disulphide redox pools, which are involved in several physiologic processes and explored as disease biomarkers. Thiol pools are regulated by mechanisms linked to their intrinsic reactivity against oxidants, concentration of antioxidants, thiol-disulphide exchange rates, and their dynamic release/removal from plasma. Major thiol couples determining plasma redox potential (Eh) are reduced cysteine (CyS)/cystine (the disulphide form of cysteine) (CySS), followed by GSH/disulphide-oxidized glutathione (GSSG). Hydrogen peroxide and hypohalous acids are the main plasma oxidants, while water-soluble and lipid-soluble small molecules are the main antioxidants. The thiol proteome and thiol-oxidoreductases are emerging investigative areas given their specific disease-related responses (e.g. protein disulphide isomerases (PDIs) in thrombosis). Plasma cysteine and glutathione redox couples exhibit pro-oxidant changes directly correlated with ageing/age-related diseases. We further discuss changes in thiol-disulphide redox state in specific groups of diseases: cardiovascular, cancer, and neurodegenerative. These results indicate association with the disease states, although not yet clear-cut to yield specific biomarkers. We also highlight mechanisms whereby thiol pools affect atherosclerosis pathophysiology. Overall, it is unlikely that a single measurement provides global assessment of plasma oxidative stress. Rather, assessment of individual thiol pools and thiol-proteins specific to any given condition has more solid and logical perspective to yield novel relevant information on disease risk and prognosis.
Collapse
|
25
|
Hydrogen Sulfide Biochemistry and Interplay with Other Gaseous Mediators in Mammalian Physiology. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:6290931. [PMID: 30050658 PMCID: PMC6040266 DOI: 10.1155/2018/6290931] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/13/2018] [Indexed: 01/06/2023]
Abstract
Hydrogen sulfide (H2S) has emerged as a relevant signaling molecule in physiology, taking its seat as a bona fide gasotransmitter akin to nitric oxide (NO) and carbon monoxide (CO). After being merely regarded as a toxic poisonous molecule, it is now recognized that mammalian cells are equipped with sophisticated enzymatic systems for H2S production and breakdown. The signaling role of H2S is mainly related to its ability to modify different protein targets, particularly by promoting persulfidation of protein cysteine residues and by interacting with metal centers, mostly hemes. H2S has been shown to regulate a myriad of cellular processes with multiple physiological consequences. As such, dysfunctional H2S metabolism is increasingly implicated in different pathologies, from cardiovascular and neurodegenerative diseases to cancer. As a highly diffusible reactive species, the intra- and extracellular levels of H2S have to be kept under tight control and, accordingly, regulation of H2S metabolism occurs at different levels. Interestingly, even though H2S, NO, and CO have similar modes of action and parallel regulatory targets or precisely because of that, there is increasing evidence of a crosstalk between the three gasotransmitters. Herein are reviewed the biochemistry, metabolism, and signaling function of hydrogen sulfide, as well as its interplay with the other gasotransmitters, NO and CO.
Collapse
|
26
|
Olson KR. H 2S and polysulfide metabolism: Conventional and unconventional pathways. Biochem Pharmacol 2017; 149:77-90. [PMID: 29248597 DOI: 10.1016/j.bcp.2017.12.010] [Citation(s) in RCA: 105] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2017] [Accepted: 12/12/2017] [Indexed: 12/13/2022]
Abstract
It is now well established that hydrogen sulfide (H2S) is an effector of a wide variety of physiological processes. It is also clear that many of the effects of H2S are mediated through reactions with cysteine sulfur on regulatory proteins and most of these are not mediated directly by H2S but require prior oxidation of H2S and the formation of per- and polysulfides (H2Sn, n = 2-8). Attendant with understanding the regulatory functions of H2S and H2Sn is an appreciation of the mechanisms that control, i.e., both increase and decrease, their production and catabolism. Although a number of standard "conventional" pathways have been described and well characterized, novel "unconventional" pathways are continuously being identified. This review summarizes our current knowledge of both the conventional and unconventional.
Collapse
Affiliation(s)
- Kenneth R Olson
- Indiana University School of Medicine - South Bend, South Bend, IN 46617, USA.
| |
Collapse
|
27
|
Caussé E, Fournier P, Roncalli J, Salvayre R, Galinier M. Serum allantoin and aminothiols as biomarkers of chronic heart failure. Acta Cardiol 2017; 72:397-403. [PMID: 28705092 DOI: 10.1080/00015385.2017.1335104] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
Background Oxidative stress (OS) represents the primary mediator of chronic heart failure (CHF) development and progression. It is well established that homocysteine is able to generate reactive oxygen species. Small amounts of allantoin in human serum result from free radical action on urate and may provide a stable marker for in vivo free radical activity. To investigate whether some easily measurable indexes such as antioxidants (uric acid, glutathione) and related molecules (allantoin, homocysteine and cysteine) can serve as OS biomarkers. Methods We investigated 75 stable CHF patients. Aminothiols and purine compound levels were determined by capillary electrophoresis. Results The homocysteine level was markedly elevated in CHF patients, whatever the aetiology. Parameters of the transsulfuration pathway and the investigated purine compounds were significantly increased. Conversely, total glutathione was decreased. The allantoin/uric acid ratio was significantly higher in CHF patients with an hyperhomocysteinaemia >17 μmol/L. All parameters of the transsulfuration and purine degadation pathways were significantly correlated, suggesting an OS in CHF patients. Conclusion Our data show an imbalance of serum aminothiols and purine compounds in these CHF patients on adapted therapy. We suggest that the evaluation and control of these new markers may help improve the OS that participates in the progression of the disease.
Collapse
Affiliation(s)
- Elizabeth Caussé
- Biochemistry Laboratory, Rangueil University Hospital, Toulouse, France
| | - Pauline Fournier
- Department of Cardiology, Rangueil University Hospital, Toulouse, France
| | - Jérome Roncalli
- Department of Cardiology, Rangueil University Hospital, Toulouse, France
- INSERM UMR 1048, Toulouse, France
| | - Robert Salvayre
- Biochemistry Laboratory, Rangueil University Hospital, Toulouse, France
- INSERM UMR 1048, Toulouse, France
| | - Michel Galinier
- Department of Cardiology, Rangueil University Hospital, Toulouse, France
- INSERM UMR 1048, Toulouse, France
| |
Collapse
|
28
|
Donnarumma E, Trivedi RK, Lefer DJ. Protective Actions of H2S in Acute Myocardial Infarction and Heart Failure. Compr Physiol 2017; 7:583-602. [PMID: 28333381 DOI: 10.1002/cphy.c160023] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Hydrogen sulfide (H2S) was identified as the third gasotransmitter in 1996 following the discoveries of the biological importance of nitric oxide and carbon monoxide. Although H2S has long been considered a highly toxic gas, the discovery of its presence and enzymatic production in mammalian tissues supports a critical role for this physiological signaling molecule. H2S is synthesized endogenously by three enzymes: cystathionine β-synthase, cystathionine-γ-lyase, and 3-mercaptopyruvate sulfurtransferase. H2S plays a pivotal role in the regulation of cardiovascular function as H2S has been shown to modulate: vasodilation, angiogenesis, inflammation, oxidative stress, and apoptosis. Perturbation of endogenous production of H2S has been associated with many pathological conditions of the cardiovascular system such as diabetes, heart failure, and hypertension. As such, modulation of the endogenous H2S signaling pathway or administration of exogenous H2S has been shown to be cytoprotective. This review article will provide a summary of the current body of evidence on the role of H2S signaling in the setting of myocardial ischemia and heart failure. © 2017 American Physiological Society. Compr Physiol 7:583-602, 2017.
Collapse
Affiliation(s)
- Erminia Donnarumma
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - Rishi K Trivedi
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| | - David J Lefer
- Cardiovascular Center of Excellence Louisiana State University Health Sciences Center, New Orleans, Louisiana, USA
| |
Collapse
|
29
|
The Sulfur Metabolite Lanthionine: Evidence for a Role as a Novel Uremic Toxin. Toxins (Basel) 2017; 9:toxins9010026. [PMID: 28075397 PMCID: PMC5308258 DOI: 10.3390/toxins9010026] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/28/2016] [Accepted: 01/02/2017] [Indexed: 12/31/2022] Open
Abstract
Lanthionine is a nonproteinogenic amino acid, composed of two alanine residues that are crosslinked on their β-carbon atoms by a thioether linkage. It is biosynthesized from the condensation of two cysteine molecules, while the related compound homolanthionine is formed from the condensation of two homocysteine molecules. The reactions can be carried out by either cystathionine-β-synthase (CBS) or cystathionine-γ-lyase (CSE) independently, in the alternate reactions of the transsulfuration pathway devoted to hydrogen sulfide biosynthesis. Low plasma total hydrogen sulfide levels, probably due to reduced CSE expression, are present in uremia, while homolanthionine and lanthionine accumulate in blood, the latter several fold. Uremic patients display a derangement of sulfur amino acid metabolism with a high prevalence of hyperhomocysteinemia. Uremia is associated with a high cardiovascular mortality, the causes of which are still not completely explained, but are related to uremic toxicity, due to the accumulation of retention products. Lanthionine inhibits hydrogen sulfide production in hepatoma cells, possibly through CBS inhibition, thus providing some basis for the biochemical mechanism, which may significantly contribute to alterations of metabolism sulfur compounds in these subjects (e.g., high homocysteine and low hydrogen sulfide). We therefore suggest that lanthionine is a novel uremic toxin.
Collapse
|
30
|
Stojanovic M, Zivkovic V, Srejovic I, Jakovljevic V, Jeremic N, Djuric D. The role of hydrogen sulfide in homocysteine-induced cardiodynamic effects and oxidative stress markers in the isolated rat heart. Physiol Int 2016; 103:428-438. [DOI: 10.1556/2060.103.2016.4.3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
This study aimed to assess the role of H2S in homocysteine-induced cardiodynamic effects in the isolated rat heart. The hearts were retrogradely perfused according to the Langendorff technique. The maximum and minimum rates of pressure in the left ventricle (dp/dt max, dp/dt min), systolic and diastolic left ventricular pressures (SLVP, DLVP), heart rate (HR), and coronary flow (CF) were measured. A spectrophotometrical method was used to measure the following oxidative stress markers: index of lipid peroxidation (thiobarbituric acid reactive substances, TBARS), nitrite level (NO2−), superoxide anion radicals (O2•−), and hydrogen peroxide (H2O2) concentrations. The administration of 10 µmol/l DL-homocysteine (DL-Hcy) alone decreased dp/dt max, SLVP, and CF but did not change any oxidative stress parameters. The administration of 10 µmol/l DL-propargylglycine (DL-PAG) decreased all cardiodynamic parameters and increased the concentration of O2•−. The co-administration of DL-Hcy and DL-PAG induced a significant decrease in all estimated cardiodynamic parameters and decreased the concentration of NO2− and O2•− but increased the levels of TBARS and H2O2. Homocysteine shows a lower pro-oxidative effect in the presence of hydrogen sulfide (H2S), which indicates a potential anti-oxidative capacity of H2S.
Collapse
Affiliation(s)
- M Stojanovic
- 1 Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - V Zivkovic
- 1 Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - I Srejovic
- 1 Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - V Jakovljevic
- 1 Department of Physiology, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - N Jeremic
- 2 Department of Pharmacy, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - D Djuric
- 3 Institute of Medical Physiology “Richard Burian”, School of Medicine, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
31
|
Yuan S, Kevil CG. Nitric Oxide and Hydrogen Sulfide Regulation of Ischemic Vascular Remodeling. Microcirculation 2016; 23:134-45. [PMID: 26381654 DOI: 10.1111/micc.12248] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 09/13/2015] [Indexed: 12/22/2022]
Abstract
Blockage or restriction of blood flow through conduit arteries results in tissue ischemia downstream of the disturbed area. Local tissues can adapt to this challenge by stimulating vascular remodeling through angiogenesis and arteriogenesis thereby restoring blood perfusion and removal of wastes. Multiple molecular mechanisms of vascular remodeling during ischemia have been identified and extensively studied. However, therapeutic benefits from these findings and insights are limited due to the complexity of various signaling networks and a lack of understanding central metabolic regulators governing these responses. The gasotransmitters NO and H2 S have emerged as master regulators that influence multiple molecular targets necessary for ischemic vascular remodeling. In this review, we discuss how NO and H2 S are individually regulated under ischemia, what their roles are in angiogenesis and arteriogenesis, and how their interaction controls ischemic vascular remodeling.
Collapse
Affiliation(s)
- Shuai Yuan
- Departments of Pathology, Molecular and Cellular Physiology, and Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana, USA
| | - Christopher G Kevil
- Departments of Pathology, Molecular and Cellular Physiology, and Cell Biology and Anatomy, LSU Health Shreveport, Shreveport, Louisiana, USA
| |
Collapse
|
32
|
Bruzzese L, Fenouillet E, Fromonot J, Durand-Gorde JM, Condo J, Kipson N, Mottola G, Deharo P, Guieu R, Ruf J. High homocysteine levels prevent via H2 S the CoCl2 -induced alteration of lymphocyte viability. J Cell Mol Med 2016; 20:1411-9. [PMID: 27061011 PMCID: PMC4956953 DOI: 10.1111/jcmm.12829] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/05/2016] [Indexed: 12/21/2022] Open
Abstract
High homocysteine (HCy) levels are associated with lymphocyte‐mediated inflammatory responses that are sometimes in turn related to hypoxia. Because adenosine is a potent lymphocyte suppressor produced in hypoxic conditions and shares metabolic pathways with HCy, we addressed the influence of high HCy levels on the hypoxia‐induced, adenosine‐mediated, alteration of lymphocyte viability. We treated mitogen‐stimulated human lymphocytes isolated from healthy individuals and the human lymphoma T‐cell line CEM with cobalt chloride (CoCl2)to reproduce hypoxia. We found that CoCl2‐altered cell viability was dose‐dependently reversed using HCy. In turn, the HCy effect was inhibited using DL‐propargylglycine, a specific inhibitor of the hydrogen sulphide (H2S)‐synthesizing enzyme cystathionine‐γ‐lyase involved in HCy catabolism. We then addressed the intracellular metabolic pathway of adenosine and HCy, and the role of the adenosine A2A receptor (A2AR). We observed that: (i) hypoxic conditions lowered the intracellular concentration of HCy by increasing adenosine production, which resulted in high A2AR expression and 3′, 5′‐cyclic adenosine monophosphate production; (ii) increasing intracellular HCy concentration reversed the hypoxia‐induced adenosinergic signalling despite high adenosine concentration by promoting both S‐adenosylhomocysteine and H2S production; (iii) DL‐propargylglycine that inhibits H2S production abolished the HCy effect. Together, these data suggest that high HCy levels prevent, via H2S production and the resulting down‐regulation of A2AR expression, the hypoxia‐induced adenosinergic alteration of lymphocyte viability. We point out the relevance of these mechanisms in the pathophysiology of cardiovascular diseases.
Collapse
Affiliation(s)
- Laurie Bruzzese
- UMR-MD2, Marseille Medical School, Aix-Marseille University and IRBA, Northern sector, Marseille, France
| | - Emmanuel Fenouillet
- UMR-MD2, Marseille Medical School, Aix-Marseille University and IRBA, Northern sector, Marseille, France.,CNRS, Institut des Sciences Biologiques, France
| | - Julien Fromonot
- UMR-MD2, Marseille Medical School, Aix-Marseille University and IRBA, Northern sector, Marseille, France.,Laboratory of Biochemistry, Timone University Hospital, Marseille, France
| | - Josée-Martine Durand-Gorde
- UMR-MD2, Marseille Medical School, Aix-Marseille University and IRBA, Northern sector, Marseille, France
| | - Jocelyne Condo
- UMR-MD2, Marseille Medical School, Aix-Marseille University and IRBA, Northern sector, Marseille, France
| | - Nathalie Kipson
- UMR-MD2, Marseille Medical School, Aix-Marseille University and IRBA, Northern sector, Marseille, France
| | - Giovanna Mottola
- UMR-MD2, Marseille Medical School, Aix-Marseille University and IRBA, Northern sector, Marseille, France.,Laboratory of Biochemistry, Timone University Hospital, Marseille, France
| | - Pierre Deharo
- UMR-MD2, Marseille Medical School, Aix-Marseille University and IRBA, Northern sector, Marseille, France.,Cardiology Department, Timone University Hospital, Marseille, France
| | - Régis Guieu
- UMR-MD2, Marseille Medical School, Aix-Marseille University and IRBA, Northern sector, Marseille, France.,Laboratory of Biochemistry, Timone University Hospital, Marseille, France
| | - Jean Ruf
- UMR-MD2, Marseille Medical School, Aix-Marseille University and IRBA, Northern sector, Marseille, France.,INSERM, France
| |
Collapse
|
33
|
Vicente JB, Malagrinò F, Arese M, Forte E, Sarti P, Giuffrè A. Bioenergetic relevance of hydrogen sulfide and the interplay between gasotransmitters at human cystathionine β-synthase. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:1127-1138. [PMID: 27039165 DOI: 10.1016/j.bbabio.2016.03.030] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Revised: 03/07/2016] [Accepted: 03/28/2016] [Indexed: 12/27/2022]
Abstract
Merely considered as a toxic gas in the past, hydrogen sulfide (H2S) is currently viewed as the third 'gasotransmitter' in addition to nitric oxide (NO) and carbon monoxide (CO), playing a key signalling role in human (patho)physiology. H2S can either act as a substrate or, similarly to CO and NO, an inhibitor of mitochondrial respiration, in the latter case by targeting cytochrome c oxidase (CcOX). The impact of H(2)S on mitochondrial energy metabolism crucially depends on the bioavailability of this gaseous molecule and its interplay with the other two gasotransmitters. The H(2)S-producing human enzyme cystathionine β-synthase (CBS), sustaining cellular bioenergetics in colorectal cancer cells, plays a role in the interplay between gasotransmitters. The enzyme was indeed recently shown to be negatively modulated by physiological concentrations of CO and NO, particularly in the presence of its allosteric activator S-adenosyl-l-methionine (AdoMet). These newly discovered regulatory mechanisms are herein reviewed. This article is part of a Special Issue entitled 'EBEC 2016: 19th European Bioenergetics Conference, Riva del Garda, Italy, July 2-6, 2016', edited by Prof. Paolo Bernardi.
Collapse
Affiliation(s)
- João B Vicente
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Av. da República (EAN), 2780-156 Oeiras, Portugal.
| | - Francesca Malagrinò
- Department of Biochemical Sciences and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Marzia Arese
- Department of Biochemical Sciences and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Elena Forte
- Department of Biochemical Sciences and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Paolo Sarti
- Department of Biochemical Sciences and Istituto Pasteur-Fondazione Cenci Bolognetti, Sapienza University of Rome, Piazzale Aldo Moro 5, I-00185 Rome, Italy
| | - Alessandro Giuffrè
- CNR Institute of Molecular Biology and Pathology, Piazzale Aldo Moro 5, I-00185 Rome, Italy.
| |
Collapse
|
34
|
Mayo JN, Chen CH, Liao FF, Bearden SE. Homocysteine disrupts outgrowth of microvascular endothelium by an iNOS-dependent mechanism. Microcirculation 2015; 21:541-50. [PMID: 24655004 DOI: 10.1111/micc.12133] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 03/17/2014] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To test the hypothesis that Hcy impairs angiogenic outgrowth through an iNOS-dependent mechanism. METHODS Adult C57Bl/6 mouse choroid explants were used in angiogenic outgrowth assays. Mouse microvascular endothelial cells were studied in culture during scrape-induced migration and dispersed cell locomotion experiments. Activity of iNOS was manipulated with pharmacology (1400 W), siRNA, and by use of choroid explants from iNOS knockout mice (iNOS(-/-)). RESULTS Hcy (20 μM) significantly decreased the area of endothelial outgrowth without altering the number of cells in the choroid explant angiogenic assay, resulting in more densely packed outgrowth. Hcy prevented the outward orientation of actin filaments and decreased the number of actin projections along the leading edge of outgrowth. Hcy also slowed outgrowth from the edge of a scraped endothelial monolayer and in cultures of dispersed cells, Hcy impaired cell locomotion without affecting proliferation. Inhibition of iNOS activity rescued the effect of Hcy on area of explant outgrowth, cell density, number of projections, cell locomotion, and rate of outgrowth following scraping. CONCLUSIONS Hcy impairs microvascular endothelial outgrowth, but not proliferation, by disrupting cell locomotion through an iNOS-dependent mechanism.
Collapse
Affiliation(s)
- Jamie N Mayo
- Department of Biological Sciences, Idaho State University, Pocatello, Idaho, USA
| | | | | | | |
Collapse
|
35
|
Abstract
SIGNIFICANCE Although oxygen (O2)-sensing cells and tissues have been known for decades, the identity of the O2-sensing mechanism has remained elusive. Evidence is accumulating that O2-dependent metabolism of hydrogen sulfide (H2S) is this enigmatic O2 sensor. RECENT ADVANCES The elucidation of biochemical pathways involved in H2S synthesis and metabolism have shown that reciprocal H2S/O2 interactions have been inexorably linked throughout eukaryotic evolution; there are multiple foci by which O2 controls H2S inactivation, and the effects of H2S on downstream signaling events are consistent with those activated by hypoxia. H2S-mediated O2 sensing has been demonstrated in a variety of O2-sensing tissues in vertebrate cardiovascular and respiratory systems, including smooth muscle in systemic and respiratory blood vessels and airways, carotid body, adrenal medulla, and other peripheral as well as central chemoreceptors. CRITICAL ISSUES Information is now needed on the intracellular location and stoichometry of these signaling processes and how and which downstream effectors are activated by H2S and its metabolites. FUTURE DIRECTIONS Development of specific inhibitors of H2S metabolism and effector activation as well as cellular organelle-targeted compounds that release H2S in a time- or environmentally controlled way will not only enhance our understanding of this signaling process but also provide direction for future therapeutic applications.
Collapse
Affiliation(s)
- Kenneth R Olson
- Department of Physiology, Indiana University School of Medicine-South Bend , South Bend, India na
| |
Collapse
|
36
|
Revsbech IG, Shen X, Chakravarti R, Jensen FB, Thiel B, Evans AL, Kindberg J, Fröbert O, Stuehr DJ, Kevil CG, Fago A. Hydrogen sulfide and nitric oxide metabolites in the blood of free-ranging brown bears and their potential roles in hibernation. Free Radic Biol Med 2014; 73:349-57. [PMID: 24909614 PMCID: PMC4413933 DOI: 10.1016/j.freeradbiomed.2014.05.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2014] [Revised: 05/19/2014] [Accepted: 05/30/2014] [Indexed: 02/08/2023]
Abstract
During winter hibernation, brown bears (Ursus arctos) lie in dens for half a year without eating while their basal metabolism is largely suppressed. To understand the underlying mechanisms of metabolic depression in hibernation, we measured type and content of blood metabolites of two ubiquitous inhibitors of mitochondrial respiration, hydrogen sulfide (H2S) and nitric oxide (NO), in winter-hibernating and summer-active free-ranging Scandinavian brown bears. We found that levels of sulfide metabolites were overall similar in summer-active and hibernating bears but their composition in the plasma differed significantly, with a decrease in bound sulfane sulfur in hibernation. High levels of unbound free sulfide correlated with high levels of cysteine (Cys) and with low levels of bound sulfane sulfur, indicating that during hibernation H2S, in addition to being formed enzymatically from the substrate Cys, may also be regenerated from its oxidation products, including thiosulfate and polysulfides. In the absence of any dietary intake, this shift in the mode of H2S synthesis would help preserve free Cys for synthesis of glutathione (GSH), a major antioxidant found at high levels in the red blood cells of hibernating bears. In contrast, circulating nitrite and erythrocytic S-nitrosation of glyceraldehyde-3-phosphate dehydrogenase, taken as markers of NO metabolism, did not change appreciably. Our findings reveal that remodeling of H2S metabolism and enhanced intracellular GSH levels are hallmarks of the aerobic metabolic suppression of hibernating bears.
Collapse
Affiliation(s)
- Inge G Revsbech
- Department of Bioscience, Aarhus University, 8000 Aarhus C, Denmark
| | - Xinggui Shen
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Ritu Chakravarti
- Department of Pathobiology, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Frank B Jensen
- Department of Biology, University of Southern Denmark, 5230 Odense M, Denmark
| | - Bonnie Thiel
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Alina L Evans
- Department of Forestry and Wildlife Management, Faculty of Applied Ecology and Agricultural Sciences, Hedmark University College, Campus Evenstad, 2418 Elverum, Norway
| | - Jonas Kindberg
- Department of Wildlife, Fish, and Environmental Studies, Swedish University of Agricultural Sciences, 90183 Umeå, Sweden
| | - Ole Fröbert
- Department of Cardiology, Örebro University Hospital, 70362 Örebro, Sweden
| | - Dennis J Stuehr
- Department of Pathobiology, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA
| | - Christopher G Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, LA 71130, USA
| | - Angela Fago
- Department of Bioscience, Aarhus University, 8000 Aarhus C, Denmark.
| |
Collapse
|
37
|
Homocysteine in renovascular complications: hydrogen sulfide is a modulator and plausible anaerobic ATP generator. Nitric Oxide 2014; 41:27-37. [PMID: 24963795 DOI: 10.1016/j.niox.2014.06.006] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Revised: 06/16/2014] [Accepted: 06/17/2014] [Indexed: 11/23/2022]
Abstract
Homocysteine (Hcy) is a non-protein amino acid derived from dietary methionine. High levels of Hcy, known as hyperhomocysteinemia (HHcy) is known to cause vascular complications. In the mammalian tissue, Hcy is metabolized by transsulfuration enzymes to produce hydrogen sulfide (H2S). H2S, a pungent smelling gas was previously known for its toxic effects in the central nervous system, recent studies however has revealed protective effects in a variety of diseases including hypertension, diabetes, inflammation, atherosclerosis, and renal disease progression and failure. Interestingly, under stress conditions including hypoxia, H2S can reduce metabolic demand and also act as a substrate for ATP production. This review highlights some of the recent advances in H2S research as a potential therapeutic agent targeting renovascular diseases associated with HHcy.
Collapse
|
38
|
Billaud M, Lohman AW, Johnstone SR, Biwer LA, Mutchler S, Isakson BE. Regulation of cellular communication by signaling microdomains in the blood vessel wall. Pharmacol Rev 2014; 66:513-69. [PMID: 24671377 PMCID: PMC3973613 DOI: 10.1124/pr.112.007351] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
It has become increasingly clear that the accumulation of proteins in specific regions of the plasma membrane can facilitate cellular communication. These regions, termed signaling microdomains, are found throughout the blood vessel wall where cellular communication, both within and between cell types, must be tightly regulated to maintain proper vascular function. We will define a cellular signaling microdomain and apply this definition to the plethora of means by which cellular communication has been hypothesized to occur in the blood vessel wall. To that end, we make a case for three broad areas of cellular communication where signaling microdomains could play an important role: 1) paracrine release of free radicals and gaseous molecules such as nitric oxide and reactive oxygen species; 2) role of ion channels including gap junctions and potassium channels, especially those associated with the endothelium-derived hyperpolarization mediated signaling, and lastly, 3) mechanism of exocytosis that has considerable oversight by signaling microdomains, especially those associated with the release of von Willebrand factor. When summed, we believe that it is clear that the organization and regulation of signaling microdomains is an essential component to vessel wall function.
Collapse
Affiliation(s)
- Marie Billaud
- Dept. of Molecular Physiology and Biophysics, University of Virginia School of Medicine, PO Box 801394, Charlottesville, VA 22902.
| | | | | | | | | | | |
Collapse
|
39
|
LI YUNQUAN, LIU GUOHUI, CAI DIANQI, PAN BAOYING, LIN YUESE, LI XUANDI, LI SHUJUAN, ZHU LING, LIAO XINXUE, WANG HUISHEN. H2S inhibition of chemical hypoxia-induced proliferation of HPASMCs is mediated by the upregulation of COX-2/PGI2. Int J Mol Med 2013; 33:359-66. [DOI: 10.3892/ijmm.2013.1579] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2013] [Accepted: 12/02/2013] [Indexed: 11/06/2022] Open
|
40
|
Qabazard B, Ahmed S, Li L, Arlt VM, Moore PK, Stürzenbaum SR. C. elegans aging is modulated by hydrogen sulfide and the sulfhydrylase/cysteine synthase cysl-2. PLoS One 2013; 8:e80135. [PMID: 24260346 PMCID: PMC3832670 DOI: 10.1371/journal.pone.0080135] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Accepted: 09/30/2013] [Indexed: 12/28/2022] Open
Abstract
Exogenous hydrogen sulfide (H2S) administration and endogenous H2S metabolism were explored in the nematode C. elegans. Chronic treatment with a slow-releasing H2S donor, GYY4137, extended median survival by 17-23% and increased tolerance towards oxidative and endoplasmic reticulum (ER) stress. Also, cysl-2, a sulfhydrylase/cysteine synthase in C. elegans, was transcriptionally upregulated by GYY4137 treatment and the deletion of cysl-2 resulted in a significant reduction in lifespan which was partially recovered by the supplementation of GYY4137. Likewise, a mammalian cell culture system, GYY4137 was able to protect bovine aortic endothelial cells (BAECs) from oxidative stress and (H2O2)-induced cell death. Taken together, this provides further support that H2S exerts a protective function which is consistent with the longevity dividend theory. Overall, this study underlines the therapeutic potential of a slow-releasing H2S donor as regulators of the aging and cellular stress pathways.
Collapse
Affiliation(s)
- Bedoor Qabazard
- School of Biomedical Sciences, King's College London, London, United Kingdom
| | - Samanza Ahmed
- School of Biomedical Sciences, King's College London, London, United Kingdom
| | - Ling Li
- School of Biomedical Sciences, King's College London, London, United Kingdom
| | - Volker M. Arlt
- School of Biomedical Sciences, King's College London, London, United Kingdom
| | - Philip K. Moore
- Neurobiology Group, Life Sciences Institute and Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | |
Collapse
|
41
|
Phytoestrogen α-Zearalanol attenuates homocysteine-induced apoptosis in human umbilical vein endothelial cells. BIOMED RESEARCH INTERNATIONAL 2013; 2013:813450. [PMID: 24195080 PMCID: PMC3806352 DOI: 10.1155/2013/813450] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Accepted: 08/28/2013] [Indexed: 01/21/2023]
Abstract
Hyperhomocysteinemia is an independent risk factor for cardiovascular diseases. The enhanced nitrative stress plays an important role in homocysteine-induced endothelial dysfunction. Previous studies have showed that phytoestrogen α -zearalanol alleviated endothelial injury in ovariectomized hyperhomocysteinemic rats; however, the underlying mechanism remains to be clarified. This study was to investigate the effects of α -zearalanol on homocysteine-induced endothelial apoptosis in vitro and explore the possible role of nitrative stress in these effects. Results showed that homocysteine (500 μ mol/L, 24 h) induced the apoptosis of human umbilical vein endothelial cells (HUVECs) obviously, and this effect was significantly attenuated by pretreatment with α -zearalanol (10(-8)~10(-6) mol/L). Moreover, α -zearalanol downregulated proapoptotic protein Bax, upregulated antiapoptotic proteins Bcl-2 and Bcl-XL, and decreased the expression and activity of caspase-9. These findings demonstrated that α -zearalanol could effectively alleviate homocysteine-induced endothelial apoptosis, and this antiapoptosis effect might be related to the inhibition of the intrinsic pathway. Western blot indicated an enhanced 3-nitrotyrosine expression in HUVECs when challenged with homocysteine, which was attenuated by pretreatment with α -zearalanol. This result implied that inhibition of nitrative stress might play a role in the protective effect of α -zearalanol on endothelial cells. Such discovery may shed a novel light on the antiatherogenic activities of α -zearalanol in hyperhomocysteinemia.
Collapse
|
42
|
Selicharová I, Kořínek M, Demianová Z, Chrudinová M, Mládková J, Jiráček J. Effects of hyperhomocysteinemia and betaine–homocysteine S-methyltransferase inhibition on hepatocyte metabolites and the proteome. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2013; 1834:1596-606. [DOI: 10.1016/j.bbapap.2013.05.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2013] [Revised: 04/30/2013] [Accepted: 05/10/2013] [Indexed: 12/11/2022]
|
43
|
Kim JI, Choi SH, Jung KJ, Lee E, Kim HY, Park KM. Protective role of methionine sulfoxide reductase A against ischemia/reperfusion injury in mouse kidney and its involvement in the regulation of trans-sulfuration pathway. Antioxid Redox Signal 2013; 18:2241-50. [PMID: 22657153 PMCID: PMC3638512 DOI: 10.1089/ars.2012.4598] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
AIMS Methionine sulfoxide reductase A (MsrA) and methionine metabolism are associated with oxidative stress, a principal cause of ischemia/reperfusion (I/R) injury. Herein, we investigated the protective role of MsrA against kidney I/R injury and the involvement of MsrA in methionine metabolism and the trans-sulfuration pathway during I/R. RESULTS We found that MsrA gene-deleted mice (MsrA(-/-)) were more susceptible to kidney I/R injury than wild-type mice (MsrA(+/+)). Deletion of MsrA enhanced renal functional and morphological impairments, congestion, inflammatory responses, and oxidative stress under I/R conditions. Concentrations of homocysteine and H(2)S in the plasma of control MsrA(-/-) mice were significantly lower than those in control MsrA(+/+) mice. I/R reduced the levels of homocysteine and H(2)S in both MsrA(+/+) and MsrA(-/-) mice, and these reductions were significantly more profound in MsrA(-/-) than in MsrA(+/+) mice. I/R reduced the expression and activities of cystathionine-β-synthase (CBS) and cystathionine-γ-lyase (CSE), both of which are H(2)S-producing enzymes, in the kidneys. These reductions were more profound in the MsrA(-/-) mice than in the MsrA(+/+)mice. INNOVATION The data provided herein constitute the first in vivo evidence for the involvement of MsrA in regulating methionine metabolism and the trans-sulfuration pathway under normal and I/R conditions. CONCLUSION Our data demonstrate that MsrA protects the kidney against I/R injury, and that this protection is associated with reduced oxidative stress and inflammatory responses. The data indicate that MsrA regulates H(2)S production during I/R by modulating the expression and activity of the CBS and CSE enzymes.
Collapse
Affiliation(s)
- Jee In Kim
- Department of Anatomy, Kyungpook National University School of Medicine, Daegu, Republic of Korea
| | | | | | | | | | | |
Collapse
|
44
|
Biology and therapeutic potential of hydrogen sulfide and hydrogen sulfide-releasing chimeras. Biochem Pharmacol 2012; 85:689-703. [PMID: 23103569 DOI: 10.1016/j.bcp.2012.10.019] [Citation(s) in RCA: 230] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 10/19/2012] [Accepted: 10/19/2012] [Indexed: 12/20/2022]
Abstract
Hydrogen sulfide, H2S, is a colorless gas with a strong odor that until recently was only considered to be a toxic environmental pollutant with little or no physiological significance. However, the past few years have demonstrated its role in many biological systems and it is becoming increasingly clear that H2S is likely to join nitric oxide (NO) and carbon monoxide (CO) as a major player in mammalian biology. In this review, we have provided an overview of the chemistry and biology of H2S and have summarized the chemistry and biological activity of some natural and synthetic H2S-donating compounds. The naturally occurring compounds discussed include, garlic, sulforaphane, erucin, and iberin. The synthetic H2S donors reviewed include, GYY4137; cysteine analogs; S-propyl cysteine, S-allyl cysteine, S-propargyl cysteine, and N-acetyl cysteine. Dithiolethione and its NSAID and other chimeras such as, L-DOPA, sildenafil, aspirin, diclofenac, naproxen, ibuprofen, indomethacin, and mesalamine have also been reviewed in detail. The newly reported NOSH-aspirin that releases both NO and H2S has also been discussed.
Collapse
|
45
|
Olson KR. A practical look at the chemistry and biology of hydrogen sulfide. Antioxid Redox Signal 2012; 17:32-44. [PMID: 22074253 PMCID: PMC3342559 DOI: 10.1089/ars.2011.4401] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2011] [Revised: 11/10/2011] [Accepted: 11/11/2011] [Indexed: 12/13/2022]
Abstract
SIGNIFICANCE Hydrogen sulfide (H(2)S) is garnering increasing interest as a biologically relevant signaling molecule. The effects of H(2)S have now been observed in virtually every organ system and numerous physiological processes. RECENT ADVANCES These studies have not only opened a new field of "gasotransmitter" biology, they have also led to the development of synthetic H(2)S "donating" compounds with the potential to be parlayed into a variety of therapeutic applications. CRITICAL ISSUES Often lost in the exuberance of this new field is a critical examination or understanding of practical aspects of H(2)S chemistry and biology. This is especially notable in the areas of handling and measuring H(2)S, evaluating biosynthetic and metabolic pathways, and separating physiological from pharmacological responses. FUTURE DIRECTIONS This brief review describes some of the pitfalls in H(2)S chemistry and biology that can lead or have already led to misleading or erroneous conclusions. The intent is to allow individuals entering or already in this burgeoning field to critically analyze the literature and to assist them in the design of future experiments.
Collapse
Affiliation(s)
- Kenneth R Olson
- Indiana University School of Medicine - South Bend, Raclin Charmichael Hall, 1234 Notre Dame Avenue, South Bend, IN 46617, USA.
| |
Collapse
|
46
|
Bełtowski J, Jamroz-Wiśniewska A. Modulation of h(2)s metabolism by statins: a new aspect of cardiovascular pharmacology. Antioxid Redox Signal 2012; 17:81-94. [PMID: 22034938 PMCID: PMC3342564 DOI: 10.1089/ars.2011.4358] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2011] [Accepted: 10/28/2011] [Indexed: 12/25/2022]
Abstract
SIGNIFICANCE Statins (3-hydroxy-3-methylglutarylcoenzyme A reductase inhibitors) are commonly used in the treatment of cardiovascular diseases. Statins reduce plasma low-density lipoproteins, inhibit inflammatory reaction, improve endothelial function, ameliorate oxidative stress, and reduce platelet activity. Consequently, statins markedly decrease the risk of acute cardiovascular events. H(2)S is synthesized in all layers of the vascular wall, including the endothelium, smooth muscle cells, and perivascular adipose tissue (PVAT). RECENT ADVANCES Recent studies demonstrate that PVAT-derived H(2)S decreases vascular tone by activating K(ATP) and/or KCNQ potassium channels in smooth muscle cells. Lipophilic atorvastatin, but not hydrophilic pravastatin, increases net H(2)S production in PVAT by inhibiting its mitochondrial oxidation, and augments the anticontractile effect of PVAT. Inhibition of H(2)S metabolism results from atorvastatin-induced decrease in coenzyme Q, which is a cofactor of H(2)S oxidation by sulfide:quinone oxidoreductase. In contrast to H(2)S, statins do not impair mitochondrial oxidation of organic substrates. CRITICAL ISSUES Taking into account antiatherosclerotic and anti-inflammatory effect of H(2)S, the gas may mediate some of the beneficial effects of statins on the cardiovascular system. In addition, specific statins differ in their ability to enhance H(2)S signaling. FUTURE DIRECTIONS Since both statins and H(2)S reduce ischemia-reperfusion injury, the possible effect of statins on H(2)S oxidation in other tissues such as the heart and the kidney needs to be examined. Inhibition of H(2)S metabolism may be a new therapeutic strategy to improve H(2)S signaling, especially in the mitochondrial compartment.
Collapse
Affiliation(s)
- Jerzy Bełtowski
- Department of Pathophysiology, Medical University, Lublin, Poland.
| | | |
Collapse
|
47
|
Mayo JN, Beard RS, Price TO, Chen CH, Erickson MA, Ercal N, Banks WA, Bearden SE. Nitrative stress in cerebral endothelium is mediated by mGluR5 in hyperhomocysteinemia. J Cereb Blood Flow Metab 2012; 32:825-34. [PMID: 22186670 PMCID: PMC3345916 DOI: 10.1038/jcbfm.2011.185] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Hyperhomocysteinemia (HHcy) disrupts nitric oxide (NO) signaling and increases nitrative stress in cerebral microvascular endothelial cells (CMVECs). This is mediated, in part, by protein nitrotyrosinylation (3-nitrotyrosine; 3-NT) though the mechanisms by which extracellular homocysteine (Hcy) generates intracellular 3-NT are unknown. Using a murine model of mild HHcy (cbs(+/-) mouse), we show that 3-NT is significantly elevated in cerebral microvessels with concomitant reductions in serum NO bioavailability as compared with wild-type littermate controls (cbs(+/+)). Directed pharmacology identified a receptor-dependent mechanism for 3-NT formation in CMVECs. Homocysteine increased expression of inducible NO synthase (iNOS) and formation of 3-NT, both of which were blocked by inhibition of metabotropic glutamate receptor-5 (mGluR5) with the specific antagonist 2-methyl-6-(phenylethynyl) pyridine hydrochloride. Activation of mGluR5 is both sufficient and necessary to drive the nitrative stress because direct activation using the mGluR5-specific agonist (RS)-2-chloro-5-hydroxyphenylglycine also increased iNOS expression and 3-NT formation while knockdown of mGluR5 receptor expression by short hairpin RNA (shRNA) blocked their increase in response to Hcy. Nitric oxide derived from iNOS was required for Hcy-mediated formation of 3-NT because the effect was blocked by 1400W. These results provide the first evidence for a receptor-dependent process that explains how plasma Hcy levels control intracellular nitrative stress in cerebral microvascular endothelium.
Collapse
Affiliation(s)
- Jamie N Mayo
- Department of Biological Sciences, Idaho State University, Pocatello, ID 83209, USA
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Metabotropic glutamate receptor 5 mediates phosphorylation of vascular endothelial cadherin and nuclear localization of β-catenin in response to homocysteine. Vascul Pharmacol 2012; 56:159-67. [PMID: 22285407 DOI: 10.1016/j.vph.2012.01.004] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2011] [Revised: 12/31/2011] [Accepted: 01/16/2012] [Indexed: 11/22/2022]
Abstract
Elevated plasma homocysteine (Hcy) is an independent risk factor for vascular disease and stroke in part by causing generalized endothelial dysfunction. A receptor that is sensitive to Hcy and its intracellular signaling systems has not been identified. β-catenin is a pleiotropic regulator of transcription and cell function. Using a brain microvascular endothelial cell line (bEnd.3), we tested the hypothesis that Hcy causes receptor-dependent nuclear translocation of β-catenin. Hcy increased phosphorylation of Y731 on vascular endothelial cadherin (VE-cadherin), a site involved in coupling β-catenin to VE-cadherin. This was blocked by inhibition of either metabotropic glutamate receptor 5 (mGluR5) or ionotropic glutamate receptor (NMDAr) and by shRNA knockdown of mGluR5. Expression of these receptors was confirmed by flow cytometry, immunohistochemistry, and western blotting. Directed pharmacology with specific agonists elucidated a signaling cascade where Hcy activates mGluR5 which activates NMDAr with subsequent PKC activation and uncoupling of the VE-cadherin/β-catenin complex. Moreover, Hcy caused a shift in localization of β-catenin from membrane-bound VE-cadherin to the cell nucleus, where it bound DNA, including a regulatory region of the gene for claudin-5, leading to reduced expression of claudin-5. Nuclear localization, DNA binding of β-catenin, and reduced claudin-5 expression were blocked by inhibition of mGluR5. Knockdown of mGluR5 expression with shRNA also rescued claudin-5 expression from the effects of Hcy treatment. These data uniquely identify mGluR5 as a master switch that drives β-catenin nuclear localization in vascular endothelium and regulates cell-cell coupling in response to elevated Hcy levels. These studies dissect a pharmacological opportunity for developing new therapeutic strategies in HHcy.
Collapse
|
49
|
Elshorbagy AK, Kozich V, Smith AD, Refsum H. Cysteine and obesity: consistency of the evidence across epidemiologic, animal and cellular studies. Curr Opin Clin Nutr Metab Care 2012; 15:49-57. [PMID: 22108094 DOI: 10.1097/mco.0b013e32834d199f] [Citation(s) in RCA: 85] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE OF REVIEW The concentrations of several plasma amino acids increase in obesity. Notably, plasma total concentrations of the sulphur amino acid cysteine (tCys) are linearly associated with fat mass in large population studies. Animal and cellular experiments support the concept that cysteine may be obesogenic. Here we review experimental and epidemiologic findings linking cysteine and related compounds with fat regulation and obesity. RECENT FINDINGS tCys, and to a lesser extent cystathionine, are the only plasma sulphur amino acids consistently associated with human obesity, whereas glutathione is inversely associated with BMI. Supplementing cyste(i)ne in rodents decreases energy expenditure and promotes adiposity, whereas defects of cysteine-synthesizing enzymes decrease body weight. In adipocytes, cysteine inhibits lipolysis and promotes lipogenesis via H2O2 production. Unlike most plasma amino acids, tCys levels do not decrease with gastric bypass-induced weight loss, further supporting the concept that elevated cysteine may be a cause, not a consequence of obesity. Although cysteine products (glutathione, taurine and H2S) are altered in obesity, they do not appear to explain cysteine's effects on body weight. SUMMARY Cellular, animal and epidemiologic data are consistent with the view that cysteine is obesogenic. Targeted research linking in-vitro and in-vivo findings is needed to elucidate mechanisms involved.
Collapse
|
50
|
Truss NJ, Warner TD. Gasotransmitters and platelets. Pharmacol Ther 2011; 132:196-203. [DOI: 10.1016/j.pharmthera.2011.07.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2011] [Accepted: 06/15/2011] [Indexed: 10/18/2022]
|