1
|
Tayama S, Kitamura Y, Hiraide K, Suzuki H, Li J, Yang Z, Mitsuwaka R, Kawajiri A, Sato K, Gao F, Nakai T, Okuyama Y, Numakura T, Yamada M, Ida T, Morita M, Kawabe T, Akaike T, Ishii N. Supersulfide controls intestinal inflammation by suppressing CD4 + T cell proliferation. Front Immunol 2025; 16:1506580. [PMID: 40303402 PMCID: PMC12037617 DOI: 10.3389/fimmu.2025.1506580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 03/25/2025] [Indexed: 05/02/2025] Open
Abstract
Inflammatory bowel disease (IBD) is characterized by chronic intestinal inflammation where CD4+ T lymphocytes play an essential role. Accumulating evidence suggests that immune responses driven by CD4+ T cells are critically regulated by various metabolic pathways including oxidative phosphorylation and glycolysis. Here we show that CARS2/CPERS-dependent supersulfide metabolism restrains CD4+ T cell proliferation in a cell-intrinsic manner. Under steady state, Cars2 +/- mice exhibited spontaneous accumulation of effector/memory CD4+ T cells in the colon with age. In lymphopenic conditions, Cars2 +/- CD4+ T cells showed enhanced cell cycle entry with reduced expression of a cell cycle inhibitor Trp53 and triggered an exacerbated form of colitis, the response being rescued by treatment with a supersulfide donor glutathione trisulfide (GSSSG). Furthermore, re-analysis of publicly available gene datasets of human colonic CD4+ T lymphocytes revealed that downregulation of CARS2 was associated with pathogenesis of IBD, and indeed, addition of GSSSG inhibited human CD4+ T cell proliferation in vitro. Together these observations reveal that CARS2/CPERS-dependent supersulfide metabolism is essential for homeostasis of intestinal effector/memory CD4+ T cells, and further suggest that dysregulation of the same metabolic pathway can lead to development of gut inflammation both in mice and humans.
Collapse
Affiliation(s)
- Shunichi Tayama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuya Kitamura
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kyoga Hiraide
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hibiki Suzuki
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Jing Li
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ziying Yang
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Ryoji Mitsuwaka
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Akihisa Kawajiri
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Kosuke Sato
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Feng Gao
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Taku Nakai
- Applied Oxygen Physiology Project, New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
- Department of Oxygen Biology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuko Okuyama
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tadahisa Numakura
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuhiro Yamada
- Department of Respiratory Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomoaki Ida
- Organization for Research Promotion, Osaka Metropolitan University, Sakai, Japan
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takeshi Kawabe
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Naoto Ishii
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
2
|
Sternberg Z. Neurodegenerative Etiology of Aromatic L-Amino Acid Decarboxylase Deficiency: a Novel Concept for Expanding Treatment Strategies. Mol Neurobiol 2024; 61:2996-3018. [PMID: 37953352 DOI: 10.1007/s12035-023-03684-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 09/29/2023] [Indexed: 11/14/2023]
Abstract
Aromatic l-amino acid decarboxylase deficiency (AADC-DY) is caused by one or more mutations in the DDC gene, resulting in the deficit in catecholamines and serotonin neurotransmitters. The disease has limited therapeutic options with relatively poor clinical outcomes. Accumulated evidence suggests the involvement of neurodegenerative mechanisms in the etiology of AADC-DY. In the absence of neurotransmitters' neuroprotective effects, the accumulation and the chronic presence of several neurotoxic metabolites including 4-dihydroxy-L-phenylalanine, 3-methyldopa, and homocysteine, in the brain of subjects with AADC-DY, promote oxidative stress and reduce the cellular antioxidant and methylation capacities, leading to glial activation and mitochondrial dysfunction, culminating to neuronal injury and death. These pathophysiological processes have the potential to hinder the clinical efficacy of treatments aimed at increasing neurotransmitters' synthesis and or function. This review describes in detail the mechanisms involved in AADC-DY neurodegenerative etiology, highlighting the close similarities with those involved in other neurodegenerative diseases. We then offer novel strategies for the treatment of the disease with the objective to either reduce the level of the metabolites or counteract their prooxidant and neurotoxic effects. These treatment modalities used singly or in combination, early in the course of the disease, will minimize neuronal injury, preserving the functional integrity of neurons, hence improving the clinical outcomes of both conventional and unconventional interventions in AADC-DY. These modalities may not be limited to AADC-DY but also to other metabolic disorders where a specific mutation leads to the accumulation of prooxidant and neurotoxic metabolites.
Collapse
Affiliation(s)
- Zohi Sternberg
- Jacobs School of Medicine and Biomedical Sciences, Buffalo Medical Center, Buffalo, NY, 14203, USA.
| |
Collapse
|
3
|
Cui J, Wang X, Dong L, Wang Q. Curcumin reduces myocardial ischemia-reperfusion injury, by increasing endogenous H 2S levels and further modulating m 6A. Mol Biol Rep 2024; 51:558. [PMID: 38643323 DOI: 10.1007/s11033-024-09478-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 03/25/2024] [Indexed: 04/22/2024]
Abstract
BACKGROUND Our previous research shows that Curcumin (CUR) attenuates myocardial ischemia-reperfusion injury (MIRI) by reducing intracellular total RNA m6A levels. However, the mechanism remains unknown. METHODS For ischemia-reperfusion (IR), H9c2 cells were cultured for 6 h in serum-free low-glycemic (1 g/L) medium and a gas environment without oxygen, and then cultured for 6 h in high-glycemic (4.5 g/L) medium supplemented with 10% FBS and a 21% oxygen environment. The effects of different concentrations of CUR (5, 10, and 20 µM) treatments on signaling molecules in conventionally cultured and IR-treated H9c2 cells were examined. RESULTS CUR treatment significantly up-regulated the H2S levels, and the mRNA and protein expression of cystathionine γ-lyase (CSE), and down-regulated the mRNAs and proteins levels of thiosulfate sulfurtransferase (TST) and ethylmalonic encephalopathy 1 (ETHE1) in H9c2 cells conventionally cultured and subjected to IR. Exogenous H2S supply (NaHS and GYY4137) significantly reduced intracellular total RNA m6A levels, and the expression of RNA m6A "writers" METTL3 and METTL14, and increased the expression of RNA m6A "eraser" FTO in H9c2 cells conventionally cultured and subjected to IR. CSE knockdown counteracted the inhibitory effect of CUR treatment on ROS production, promotion on cell viability, and inhibition on apoptosis of H9c2 cells subjected to IR. CONCLUSION CUR attenuates MIRI by regulating the expression of H2S level-regulating enzymes and increasing the endogenous H2S levels. Increased H2S levels could regulate the m6A-related proteins expression and intracellular total RNA m6A levels.
Collapse
Affiliation(s)
- Jiankun Cui
- Cardiovascular Department, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, No.26 Heping Road, Harbin, Heilongjiang, 150040, China
| | - Xin Wang
- Department of Chinese Medicine Treating Preventable Diseases, Dezhou Traditional Chinese Medicine Hospital, No. 1165 Tianqu East Road, Dezhou, 253000, China
| | - Lingling Dong
- Department of Intensive Medicine, Dezhou Traditional Chinese Medicine Hospital, No. 1165 Tianqu East Road, Dezhou, 253000, China.
| | - Qinwen Wang
- Beijing Garrison District Haidian Retired Cadres Twenty-Fifth, Beijing, 100039, China.
| |
Collapse
|
4
|
Ogata S, Matsunaga T, Jung M, Barayeu U, Morita M, Akaike T. Persulfide Biosynthesis Conserved Evolutionarily in All Organisms. Antioxid Redox Signal 2023; 39:983-999. [PMID: 37565274 PMCID: PMC10655014 DOI: 10.1089/ars.2023.0405] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/04/2023] [Indexed: 08/12/2023]
Abstract
Significance: Persulfides/polysulfides are sulfur-catenated molecular species (i.e., R-Sn-R', n > 2; R-Sn-H, n > 1, with R = cysteine, glutathione, and proteins), such as cysteine persulfide (CysSSH). These species are abundantly formed as endogenous metabolites in mammalian and human cells and tissues. However, the persulfide synthesis mechanism has yet to be thoroughly discussed. Recent Advances: We used β-(4-hydroxyphenyl)ethyl iodoacetamide and mass spectrometry to develop sulfur metabolomics, a highly precise, quantitative analytical method for sulfur metabolites. Critical Issues: With this method, we detected appreciable amounts of different persulfide species in biological specimens from various organisms, from the domains Bacteria, Archaea, and Eukarya. By using our rigorously quantitative approach, we identified cysteinyl-tRNA synthetase (CARS) as a novel persulfide synthase, and we found that the CysSSH synthase activity of CARS is highly conserved from the domains Bacteria to Eukarya. Because persulfide synthesis is found not only with CARS but also with other sulfotransferase enzymes in many organisms, persulfides/polysulfides are expected to contribute as fundamental elements to substantially diverse biological phenomena. In fact, persulfide generation in higher organisms-that is, plants and animals-demonstrated various physiological functions that are mediated by redox signaling, such as regulation of energy metabolism, infection, inflammation, and cell death, including ferroptosis. Future Directions: Investigating CARS-dependent persulfide production may clarify various pathways of redox signaling in physiological and pathophysiological conditions and may thereby promote the development of preventive and therapeutic measures for oxidative stress as well as different inflammatory, metabolic, and neurodegenerative diseases. Antioxid. Redox Signal. 39, 983-999.
Collapse
Affiliation(s)
- Seiryo Ogata
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tetsuro Matsunaga
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Minkyung Jung
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Uladzimir Barayeu
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masanobu Morita
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
5
|
Ivanov AV, Popov MA, Metelkin AA, Aleksandrin VV, Agafonov EG, Kruglova MP, Silina EV, Stupin VA, Maslennikov RA, Kubatiev AA. Influence of Coronary Artery Bypass Grafts on Blood Aminothiols in Patients with Coronary Artery Disease. Metabolites 2023; 13:743. [PMID: 37367901 PMCID: PMC10305081 DOI: 10.3390/metabo13060743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Revised: 06/03/2023] [Accepted: 06/08/2023] [Indexed: 06/28/2023] Open
Abstract
Coronary artery disease (CAD) and the coronary artery bypass graft (CABG) are associated with a decreased blood glutathione (bGSH) level. Since GSH metabolism is closely related to other aminothiols (homocysteine and cysteine) and glucose, the aim of this study was to reveal the associations of bGSH with glucose and plasma aminothiols in CAD patients (N = 35) before CABG and in the early postoperative period. Forty-three volunteers with no history of cardiovascular disease formed the control group. bGSH and its redox status were significantly lower in CAD patients at admission. CABG had no significant effect on these parameters, with the exception of an increase in the bGSH/hemoglobin ratio. At admission, CAD patients were characterized by negative associations of homocysteine and cysteine with bGSH. All these associations disappeared after CABG. An association was found between an increase in oxidized GSH in the blood in the postoperative period and fasting glucose levels. Thus, CAD is associated with the depletion of the intracellular pool and the redox status of bGSH, in which hyperhomocysteinemia and a decrease in the bioavailability of the extracellular pool of cysteine play a role. The present study indicates that CABG causes disruptions in aminothiol metabolism and induces the synthesis of bGSH. Moreover, glucose becomes an important factor in the dysregulation of GSH metabolism in CABG.
Collapse
Affiliation(s)
- Alexander Vladimirovich Ivanov
- Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia; (A.A.M.); (V.V.A.); (M.P.K.); (A.A.K.)
| | - Mikhail Aleksandrovich Popov
- Moscow Regional Research and Clinical Institute n.a. M.F. Vladimirskiy, Shchepkin St., 61/2, 129110 Moscow, Russia; (M.A.P.); (E.G.A.); (R.A.M.)
| | - Arkady Andreevich Metelkin
- Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia; (A.A.M.); (V.V.A.); (M.P.K.); (A.A.K.)
| | - Valery Vasil’evich Aleksandrin
- Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia; (A.A.M.); (V.V.A.); (M.P.K.); (A.A.K.)
| | - Evgeniy Gennad’evich Agafonov
- Moscow Regional Research and Clinical Institute n.a. M.F. Vladimirskiy, Shchepkin St., 61/2, 129110 Moscow, Russia; (M.A.P.); (E.G.A.); (R.A.M.)
| | - Maria Petrovna Kruglova
- Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia; (A.A.M.); (V.V.A.); (M.P.K.); (A.A.K.)
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya St., 8, 119991 Moscow, Russia;
| | - Ekaterina Vladimirovna Silina
- Department of Human Pathology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Trubetskaya St., 8, 119991 Moscow, Russia;
| | - Victor Aleksandrovich Stupin
- Department of Hospital Surgery No. 1, Pirogov Russian National Research Medical University, Ostrovityanova St., 1, 117997 Moscow, Russia;
| | - Ruslan Andreevich Maslennikov
- Moscow Regional Research and Clinical Institute n.a. M.F. Vladimirskiy, Shchepkin St., 61/2, 129110 Moscow, Russia; (M.A.P.); (E.G.A.); (R.A.M.)
| | - Aslan Amirkhanovich Kubatiev
- Institute of General Pathology and Pathophysiology, Baltiyskaya St., 8, 125315 Moscow, Russia; (A.A.M.); (V.V.A.); (M.P.K.); (A.A.K.)
| |
Collapse
|
6
|
Cirino G, Szabo C, Papapetropoulos A. Physiological roles of hydrogen sulfide in mammalian cells, tissues and organs. Physiol Rev 2022; 103:31-276. [DOI: 10.1152/physrev.00028.2021] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
H2S belongs to the class of molecules known as gasotransmitters, which also includes nitric oxide (NO) and carbon monoxide (CO). Three enzymes are recognized as endogenous sources of H2S in various cells and tissues: cystathionine g-lyase (CSE), cystathionine β-synthase (CBS) and 3-mercaptopyruvate sulfurtransferase (3-MST). The current article reviews the regulation of these enzymes as well as the pathways of their enzymatic and non-enzymatic degradation and elimination. The multiple interactions of H2S with other labile endogenous molecules (e.g. NO) and reactive oxygen species are also outlined. The various biological targets and signaling pathways are discussed, with special reference to H2S and oxidative posttranscriptional modification of proteins, the effect of H2S on channels and intracellular second messenger pathways, the regulation of gene transcription and translation and the regulation of cellular bioenergetics and metabolism. The pharmacological and molecular tools currently available to study H2S physiology are also reviewed, including their utility and limitations. In subsequent sections, the role of H2S in the regulation of various physiological and cellular functions is reviewed. The physiological role of H2S in various cell types and organ systems are overviewed. Finally, the role of H2S in the regulation of various organ functions is discussed as well as the characteristic bell-shaped biphasic effects of H2S. In addition, key pathophysiological aspects, debated areas, and future research and translational areas are identified A wide array of significant roles of H2S in the physiological regulation of all organ functions emerges from this review.
Collapse
Affiliation(s)
- Giuseppe Cirino
- Department of Pharmacy, School of Medicine, University of Naples Federico II, Naples, Italy
| | - Csaba Szabo
- Chair of Pharmacology, Section of Medicine, University of Fribourg, Switzerland
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece & Clinical, Experimental Surgery and Translational Research Center, Biomedical Research Foundation of the Academy of Athens, Greece
| |
Collapse
|
7
|
Hine C, Treviño-Villarreal JH, Mejia P, Longchamp A, Brace LE, Harputlugil E, Mitchell SJ, Yang J, Guan Y, Maciejewski JP, Jha BK, Mitchell JR. Sulfur Amino Acid Supplementation Abrogates Protective Effects of Caloric Restriction for Enhancing Bone Marrow Regrowth Following Ionizing Radiation. Nutrients 2022; 14:nu14071529. [PMID: 35406143 PMCID: PMC9002760 DOI: 10.3390/nu14071529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 03/30/2022] [Accepted: 04/02/2022] [Indexed: 02/01/2023] Open
Abstract
Radiation therapy damages and depletes total bone marrow (BM) cellularity, compromising safety and limiting effective dosing. Aging also strains total BM and BM hematopoietic stem and progenitor cell (HSPC) renewal and function, resulting in multi-system defects. Interventions that preserve BM and BM HSPC homeostasis thus have potential clinical significance. Here, we report that 50% calorie restriction (CR) for 7-days or fasting for 3-days prior to irradiation improved mouse BM regrowth in the days and weeks post irradiation. Specifically, one week of 50% CR ameliorated loss of total BM cellularity post irradiation compared to ad libitum-fed controls. CR-mediated BM protection was abrogated by dietary sulfur amino acid (i.e., cysteine, methionine) supplementation or pharmacological inhibition of sulfur amino acid metabolizing and hydrogen sulfide (H2S) producing enzymes. Up to 2-fold increased proliferative capacity of ex vivo-irradiated BM isolated from food restricted mice relative to control mice indicates cell autonomy of the protective effect. Pretreatment with H2S in vitro was sufficient to preserve proliferative capacity by over 50% compared to non-treated cells in ex vivo-irradiated BM and BM HSPCs. The exogenous addition of H2S inhibited Ten eleven translocation 2 (TET2) activity in vitro, thus providing a potential mechanism of action. Short-term CR or fasting therefore offers BM radioprotection and promotes regrowth in part via altered sulfur amino acid metabolism and H2S generation, with translational implications for radiation treatment and aging.
Collapse
Affiliation(s)
- Christopher Hine
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA;
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Correspondence:
| | - J. Humberto Treviño-Villarreal
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Service of Endocrinology, Department of Internal Medicine, University Hospital and School of Medicine, Universidad Autonoma de Nuevo Leon, Monterrey N.L. 64460, Mexico
| | - Pedro Mejia
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
| | - Alban Longchamp
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois, University of Lausanne, CH-1011 Lausanne, Switzerland
| | - Lear E. Brace
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
| | - Eylul Harputlugil
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
| | - Sarah J. Mitchell
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Department of Health Sciences and Technology, ETH Zurich, 8005 Zurich, Switzerland
| | - Jie Yang
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH 44195, USA;
| | - Yihong Guan
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Y.G.); (J.P.M.); (B.K.J.)
| | - Jaroslaw P. Maciejewski
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Y.G.); (J.P.M.); (B.K.J.)
| | - Babal K. Jha
- Department of Translational Hematology & Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA; (Y.G.); (J.P.M.); (B.K.J.)
| | - James R. Mitchell
- Department of Molecular Metabolism (Formally Genetics and Complex Diseases), Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; (J.H.T.-V.) (P.M.); (A.L.); (L.E.B.); (E.H.); (S.J.M.); (J.R.M.)
- Department of Health Sciences and Technology, ETH Zurich, 8005 Zurich, Switzerland
| |
Collapse
|
8
|
A High-Methionine Diet for One-Week Induces a High Accumulation of Methionine in the Cerebrospinal Fluid and Confers Bipolar Disorder-like Behavior in Mice. Int J Mol Sci 2022; 23:ijms23020928. [PMID: 35055113 PMCID: PMC8778486 DOI: 10.3390/ijms23020928] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/12/2022] [Accepted: 01/14/2022] [Indexed: 02/04/2023] Open
Abstract
Methionine (Met) is considered the most toxic amino acid in mammals. Here, we investigated biochemical and behavioral impacts of ad libitum one-week feeding of high-Met diets on mice. Adult male mice were fed the standard rodent diet that contained 0.44% Met (1×) or a diet containing 16 graded Met doses (1.2×–13×). High-Met diets for one-week induced a dose-dependent decrease in body weight and an increase in serum Met levels with a 2.55 mM peak (versus basal 53 µM) on the 12×Met diet. Total homocysteine (Hcy) levels were also upregulated while concentrations of other amino acids were almost maintained in serum. Similarly, levels of Met and Hcy (but not the other amino acids) were highly elevated in the cerebrospinal fluids of mice on the 10×Met diet; the Met levels were much higher than Hcy and the others. In a series of behavioral tests, mice on the 10×Met diet displayed increased anxiety and decreased traveled distances in an open-field test, increased activity to escape from water soaking and tail hanging, and normal learning/memory activity in a Y-maze test, which were reflections of negative/positive symptoms and normal cognitive function, respectively. These results indicate that high-Met ad libitum feeding even for a week can induce bipolar disorder-like disease models in mice.
Collapse
|
9
|
Longchamp A, MacArthur MR, Trocha K, Ganahl J, Mann CG, Kip P, King WW, Sharma G, Tao M, Mitchell SJ, Ditrói T, Yang J, Nagy P, Ozaki CK, Hine C, Mitchell JR. Plasma Hydrogen Sulfide Is Positively Associated With Post-operative Survival in Patients Undergoing Surgical Revascularization. Front Cardiovasc Med 2021; 8:750926. [PMID: 34760947 PMCID: PMC8574965 DOI: 10.3389/fcvm.2021.750926] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/13/2021] [Indexed: 12/05/2022] Open
Abstract
Objective: Hydrogen sulfide (H2S) is a gaseous signaling molecule and redox factor important for cardiovascular function. Deficiencies in its production or bioavailability are implicated in atherosclerotic disease. However, it is unknown if circulating H2S levels differ between vasculopaths and healthy individuals, and if so, whether H2S measurements can be used to predict surgical outcomes. Here, we examined: (1) Plasma H2S levels in patients undergoing vascular surgery and compared these to healthy controls, and (2) the association between H2S levels and mortality in a cohort of patients undergoing surgical revascularization. Methods: One hundred and fifteen patients undergoing carotid endarterectomy, open lower extremity revascularization or lower leg amputation were enrolled at a single institution. Peripheral blood was also collected from a matched control cohort of 20 patients without peripheral or coronary artery disease. Plasma H2S production capacity and sulfide concentration were measured using the lead acetate and monobromobimane methods, respectively. Results: Plasma H2S production capacity and plasma sulfide concentrations were reduced in patients with PAD (p < 0.001, p = 0.013, respectively). Patients that underwent surgical revascularization were divided into high vs. low H2S production capacity groups by median split. Patients in the low H2S production group had increased probability of mortality (p = 0.003). This association was robust to correction for potentially confounding variables using Cox proportional hazard models. Conclusion: Circulating H2S levels were lower in patients with atherosclerotic disease. Patients undergoing surgical revascularization with lower H2S production capacity, but not sulfide concentrations, had increased probability of mortality within 36 months post-surgery. This work provides insight on the role H2S plays as a diagnostic and potential therapeutic for cardiovascular disease.
Collapse
Affiliation(s)
- Alban Longchamp
- Department of Vascular Surgery, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland.,Department of Biomedical Sciences, University of Lausanne, Lausanne, Switzerland
| | - Michael R MacArthur
- Department of Health Sciences and Technology, Eidgenössische Technische Hochschule (ETH) Zürich, Zurich, Switzerland
| | - Kaspar Trocha
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Janine Ganahl
- Department of Molecular Metabolism, Harvard T. H. Chan School of Public Health, Boston, MA, United States
| | - Charlotte G Mann
- Department of Health Sciences and Technology, Eidgenössische Technische Hochschule (ETH) Zürich, Zurich, Switzerland
| | - Peter Kip
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - William W King
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Gaurav Sharma
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Ming Tao
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Sarah J Mitchell
- Department of Health Sciences and Technology, Eidgenössische Technische Hochschule (ETH) Zürich, Zurich, Switzerland
| | - Tamás Ditrói
- Department of Molecular Immunology and Toxicology, National Institute of Oncology, Budapest, Hungary
| | - Jie Yang
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology, National Institute of Oncology, Budapest, Hungary.,Department of Anatomy and Histology, University of Veterinary Medicine, Budapest, Hungary
| | - C Keith Ozaki
- Department of Surgery and the Heart and Vascular Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States
| | - Christopher Hine
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic Lerner Research Institute, Cleveland, OH, United States
| | - James R Mitchell
- Department of Health Sciences and Technology, Eidgenössische Technische Hochschule (ETH) Zürich, Zurich, Switzerland
| |
Collapse
|
10
|
Impact of spaceflight and artificial gravity on sulfur metabolism in mouse liver: sulfur metabolomic and transcriptomic analysis. Sci Rep 2021; 11:21786. [PMID: 34750416 PMCID: PMC8575787 DOI: 10.1038/s41598-021-01129-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 10/11/2021] [Indexed: 12/30/2022] Open
Abstract
Spaceflight induces hepatic damage, partially owing to oxidative stress caused by the space environment such as microgravity and space radiation. We examined the roles of anti-oxidative sulfur-containing compounds on hepatic damage after spaceflight. We analyzed the livers of mice on board the International Space Station for 30 days. During spaceflight, half of the mice were exposed to artificial earth gravity (1 g) using centrifugation cages. Sulfur-metabolomics of the livers of mice after spaceflight revealed a decrease in sulfur antioxidants (ergothioneine, glutathione, cysteine, taurine, thiamine, etc.) and their intermediates (cysteine sulfonic acid, hercynine, N-acethylserine, serine, etc.) compared to the controls on the ground. Furthermore, RNA-sequencing showed upregulation of gene sets related to oxidative stress and sulfur metabolism, and downregulation of gene sets related to glutathione reducibility in the livers of mice after spaceflight, compared to controls on the ground. These changes were partially mitigated by exposure to 1 g centrifugation. For the first time, we observed a decrease in sulfur antioxidants based on a comprehensive analysis of the livers of mice after spaceflight. Our data suggest that a decrease in sulfur-containing compounds owing to both microgravity and other spaceflight environments (radiation and stressors) contributes to liver damage after spaceflight.
Collapse
|
11
|
Yarmohammadi F, Hayes AW, Karimi G. The cardioprotective effects of hydrogen sulfide by targeting endoplasmic reticulum stress and the Nrf2 signaling pathway: A review. Biofactors 2021; 47:701-712. [PMID: 34161646 DOI: 10.1002/biof.1763] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 05/30/2021] [Accepted: 06/01/2021] [Indexed: 12/20/2022]
Abstract
Cardiac diseases are emerging due to lifestyle, urbanization, and the accelerated aging process. Oxidative stress has been associated with cardiac injury progression through interference with antioxidant strategies and endoplasmic reticulum (ER) function. Hydrogen sulfide (H2 S) is generated endogenously from l-cysteine in various tissues including heart tissue. Pharmacological evaluation of H2 S has suggested a potential role for H2 S against diabetic cardiomyopathy, ischemia/reperfusion injury, myocardial infarction, and cardiotoxicity. Nuclear factor E2-related factor 2 (Nrf2) activity is crucial for cell survival in response to oxidative stress. H2 S up-regulates Nrf2 expression and its related signaling pathway in myocytes. H2 S also suppresses the expression and activity of ER stress-related proteins. H2 S has been reported to improve various cardiac conditions through antioxidant and anti-ER stress-related activities.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, Florida, USA
- Institute for Integrative Toxicology, Michigan State University, East Lansing, Michigan, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
12
|
Mitochondrial Redox Signaling and Oxidative Stress in Kidney Diseases. Biomolecules 2021; 11:biom11081144. [PMID: 34439810 PMCID: PMC8391472 DOI: 10.3390/biom11081144] [Citation(s) in RCA: 109] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 07/31/2021] [Accepted: 08/01/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondria are essential organelles in physiology and kidney diseases, because they produce cellular energy required to perform their function. During mitochondrial metabolism, reactive oxygen species (ROS) are produced. ROS function as secondary messengers, inducing redox-sensitive post-translational modifications (PTM) in proteins and activating or deactivating different cell signaling pathways. However, in kidney diseases, ROS overproduction causes oxidative stress (OS), inducing mitochondrial dysfunction and altering its metabolism and dynamics. The latter processes are closely related to changes in the cell redox-sensitive signaling pathways, causing inflammation and apoptosis cell death. Although mitochondrial metabolism, ROS production, and OS have been studied in kidney diseases, the role of redox signaling pathways in mitochondria has not been addressed. This review focuses on altering the metabolism and dynamics of mitochondria through the dysregulation of redox-sensitive signaling pathways in kidney diseases.
Collapse
|
13
|
Kawano Y, Suzuki K, Ohtsu I. Development of quantitative analytical method for volatile thiol compound with LC-ESI-MS as nonvolatile derivative by integrating a thiol-specific derivatization. Biosci Biotechnol Biochem 2021; 85:1932-1936. [PMID: 34279569 DOI: 10.1093/bbb/zbab129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/06/2021] [Indexed: 11/13/2022]
Abstract
Generally, volatile thiols are hard to be measured with ESI (electrospray ionization)-type LC-MS due to the volatility. Therefore, we here evaluated the pretreatment of their S-bimanyl derivatization by monobromobimane to enable the detection as nonvolatile derivative. Consequently, we successfully developed the convenient and efficient method through the quantitative analysis of 2-furanmethanethiol (volatile thiol odorant of coffee aroma) in coffee bean.
Collapse
Affiliation(s)
- Yusuke Kawano
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.,Microbiology Research Center for Sustainability, University of Tsukuba, Tsukuba, Japan.,euglena Co., Ltd., Tokyo, Japan
| | | | - Iwao Ohtsu
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Tsukuba, Japan.,Microbiology Research Center for Sustainability, University of Tsukuba, Tsukuba, Japan.,euglena Co., Ltd., Tokyo, Japan
| |
Collapse
|
14
|
Dietary restriction transforms the mammalian protein persulfidome in a tissue-specific and cystathionine γ-lyase-dependent manner. Nat Commun 2021; 12:1745. [PMID: 33741971 PMCID: PMC7979915 DOI: 10.1038/s41467-021-22001-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Accepted: 02/24/2021] [Indexed: 02/07/2023] Open
Abstract
Hydrogen sulfide (H2S) is a cytoprotective redox-active metabolite that signals through protein persulfidation (R-SSnH). Despite the known importance of persulfidation, tissue-specific persulfidome profiles and their associated functions are not well characterized, specifically under conditions and interventions known to modulate H2S production. We hypothesize that dietary restriction (DR), which increases lifespan and can boost H2S production, expands tissue-specific persulfidomes. Here, we find protein persulfidation enriched in liver, kidney, muscle, and brain but decreased in heart of young and aged male mice under two forms of DR, with DR promoting persulfidation in numerous metabolic and aging-related pathways. Mice lacking cystathionine γ-lyase (CGL) have overall decreased tissue protein persulfidation numbers and fail to functionally augment persulfidomes in response to DR, predominantly in kidney, muscle, and brain. Here, we define tissue- and CGL-dependent persulfidomes and how diet transforms their makeup, underscoring the breadth for DR and H2S to impact biological processes and organismal health. Dietary restriction (DR) can increase protein persulfidation but the tissue specificity of this process is not well understood. Here, the authors compare organ-specific protein persulfidomes in young and aged mice under DR, and show that DR-dependent persulfidome changes depend on cystathionine γ-lyase.
Collapse
|
15
|
Casin KM, Calvert JW. Harnessing the Benefits of Endogenous Hydrogen Sulfide to Reduce Cardiovascular Disease. Antioxidants (Basel) 2021; 10:antiox10030383. [PMID: 33806545 PMCID: PMC8000539 DOI: 10.3390/antiox10030383] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 02/26/2021] [Accepted: 03/01/2021] [Indexed: 02/02/2023] Open
Abstract
Cardiovascular disease is the leading cause of death in the U.S. While various studies have shown the beneficial impact of exogenous hydrogen sulfide (H2S)-releasing drugs, few have demonstrated the influence of endogenous H2S production. Modulating the predominant enzymatic sources of H2S-cystathionine-β-synthase, cystathionine-γ-lyase, and 3-mercaptopyruvate sulfurtransferase-is an emerging and promising research area. This review frames the discussion of harnessing endogenous H2S within the context of a non-ischemic form of cardiomyopathy, termed diabetic cardiomyopathy, and heart failure. Also, we examine the current literature around therapeutic interventions, such as intermittent fasting and exercise, that stimulate H2S production.
Collapse
|
16
|
Citi V, Martelli A, Gorica E, Brogi S, Testai L, Calderone V. Role of hydrogen sulfide in endothelial dysfunction: Pathophysiology and therapeutic approaches. J Adv Res 2021; 27:99-113. [PMID: 33318870 PMCID: PMC7728589 DOI: 10.1016/j.jare.2020.05.015] [Citation(s) in RCA: 65] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 05/12/2020] [Accepted: 05/13/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND The vascular endothelium represents a fundamental mechanical and biological barrier for the maintenance of vascular homeostasis along the entire vascular tree. Changes in its integrity are associated to several cardiovascular diseases, including hypertension, atherosclerosis, hyperhomocysteinemia, diabetes, all linked to the peculiar condition named endothelial dysfunction, which is referred to the loss of endothelial physiological functions, comprehending the regulation of vascular relaxation and/or cell redox balance, the inhibition of leukocyte infiltration and the production of NO. Among the endothelium-released vasoactive factors, in the last years hydrogen sulfide has been viewed as one of the main characters involved in the regulation of endothelium functionality, and many studies demonstrated that H2S behaves as a vasoprotective gasotransmitter in those cardiovascular diseases where endothelial dysfunction seems to be the central issue. AIM The role of hydrogen sulfide in endothelial dysfunction-related cardiovascular diseases is discussed in this review. KEY SCIENTIFIC CONCEPTS Possible therapeutic approaches using molecules able to release H2S.
Collapse
Affiliation(s)
- Valentina Citi
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| | - Alma Martelli
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| | - Era Gorica
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| | - Simone Brogi
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| | - Lara Testai
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| | - Vincenzo Calderone
- Department of Pharmacy, University of Pisa, via Bonanno n.6, 56125 Pisa, Italy
| |
Collapse
|
17
|
The multifaceted roles of sulfane sulfur species in cancer-associated processes. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2020; 1862:148338. [PMID: 33212042 DOI: 10.1016/j.bbabio.2020.148338] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 10/30/2020] [Accepted: 11/13/2020] [Indexed: 02/07/2023]
Abstract
Sulfane sulfur species comprise a variety of biologically relevant hydrogen sulfide (H2S)-derived species, including per- and poly-sulfidated low molecular weight compounds and proteins. A growing body of evidence suggests that H2S, currently recognized as a key signaling molecule in human physiology and pathophysiology, plays an important role in cancer biology by modulating cell bioenergetics and contributing to metabolic reprogramming. This is accomplished through functional modulation of target proteins via H2S binding to heme iron centers or H2S-mediated reversible per- or poly-sulfidation of specific cysteine residues. Since sulfane sulfur species are increasingly viewed not only as a major source of H2S but also as key mediators of some of the biological effects commonly attributed to H2S, the multifaceted role of these species in cancer biology is reviewed here with reference to H2S, focusing on their metabolism, signaling function, impact on cell bioenergetics and anti-tumoral properties.
Collapse
|
18
|
Cortassa S, Caceres V, Tocchetti CG, Bernier M, de Cabo R, Paolocci N, Sollott SJ, Aon MA. Metabolic remodelling of glucose, fatty acid and redox pathways in the heart of type 2 diabetic mice. J Physiol 2020; 598:1393-1415. [PMID: 30462352 PMCID: PMC7739175 DOI: 10.1113/jp276824] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 11/15/2018] [Indexed: 12/31/2022] Open
Abstract
KEY POINTS Hearts from type 2 diabetic animals display perturbations in excitation-contraction coupling, impairing myocyte contractility and delaying relaxation, along with altered substrate consumption patterns. Under high glucose and β-adrenergic stimulation conditions, palmitate can, at least in part, offset left ventricle (LV) dysfunction in hearts from diabetic mice, improving contractility and relaxation while restoring coronary perfusion pressure. Fluxome calculations of central catabolism in diabetic hearts show that, in the presence of palmitate, there is a metabolic remodelling involving tricarboxylic acid cycle, polyol and pentose phosphate pathways, leading to improved redox balance in cytoplasmic and mitochondrial compartments. Under high glucose and increased energy demand, the metabolic/fluxomic redirection leading to restored redox balance imparted by palmitate helps explain maintained LV function and may contribute to designing novel therapeutic approaches to prevent cardiac dysfunction in diabetic patients. ABSTRACT Type-2 diabetes (T2DM) leads to reduced myocardial performance, and eventually heart failure. Excessive accumulation of lipids and glucose is central to T2DM cardiomyopathy. Previous data showed that palmitate (Palm) or glutathione preserved heart mitochondrial energy/redox balance under excess glucose, rescuing β-adrenergic-stimulated cardiac excitation-contraction coupling. However, the mechanisms underlying the accompanying improved contractile performance have been largely ignored. Herein we explore in intact heart under substrate excess the metabolic remodelling associated with cardiac function in diabetic db/db mice subjected to stress given by β-adrenergic stimulation with isoproterenol and high glucose compared to their non-diabetic controls (+/+, WT) under euglycaemic conditions. When perfused with Palm, T2DM hearts exhibited improved contractility/relaxation compared to WT, accompanied by extensive metabolic remodelling as demonstrated by metabolomics-fluxomics combined with bioinformatics and computational modelling. The T2DM heart metabolome showed significant differences in the abundance of metabolites in pathways related to glucose, lipids and redox metabolism. Using a validated computational model of heart's central catabolism, comprising glucose and fatty acid (FA) oxidation in cytoplasmic and mitochondrial compartments, we estimated that fluxes through glucose degradation pathways are ∼2-fold lower in heart from T2DM vs. WT under all conditions studied. Palm addition elicits improvement of the redox status via enhanced β-oxidation and decreased glucose uptake, leading to flux-redirection away from redox-consuming pathways (e.g. polyol) while maintaining the flux through redox-generating pathways together with glucose-FA 'shared fuelling' of oxidative phosphorylation. Thus, available FAs such as Palm may help improve function via enhanced redox balance in T2DM hearts during peaks of hyperglycaemia and increased workload.
Collapse
Affiliation(s)
- Sonia Cortassa
- Laboratory of Cardiovascular Science, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | - Viviane Caceres
- Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, MD, 21205, USA
- Posgraduate Program in Rehabilitation Sciences, Dept. Health Sciences, Federal University of Santa Catarina, Ararangua, SC, Brazil
| | - Carlo G Tocchetti
- Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, MD, 21205, USA
- Dipartimento di Scienze Mediche Traslazionali, Universita' degli Studi di Napoli Federico II Via Pansini 5, Edificio 2, 80131, Napoli, Italy
| | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, The Johns Hopkins University, Baltimore, MD, 21205, USA
- Department of Biomedical Sciences, University of Padova, via Marzolo 3, 35131, Padova, Italy
| | - Steven J Sollott
- Laboratory of Cardiovascular Science, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
- Translational Gerontology Branch, National Institute on Aging/NIH, Baltimore, MD, 21224, USA
| |
Collapse
|
19
|
Akahoshi N, Minakawa T, Miyashita M, Sugiyama U, Saito C, Takemoto R, Honda A, Kamichatani W, Kamata S, Anan Y, Ishii I. Increased Urinary 3-Mercaptolactate Excretion and Enhanced Passive Systemic Anaphylaxis in Mice Lacking Mercaptopyruvate Sulfurtransferase, a Model of Mercaptolactate-Cysteine Disulfiduria. Int J Mol Sci 2020; 21:ijms21030818. [PMID: 32012740 PMCID: PMC7038117 DOI: 10.3390/ijms21030818] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Revised: 01/24/2020] [Accepted: 01/25/2020] [Indexed: 12/19/2022] Open
Abstract
Mercaptopyruvate sulfurtransferase (Mpst) and its homolog thiosulfate sulfurtransferase (Tst = rhodanese) detoxify cyanide to thiocyanate. Mpst is attracting attention as one of the four endogenous hydrogen sulfide (H2S)/reactive sulfur species (RSS)-producing enzymes, along with cystathionine β-synthase (Cbs), cystathionine γ-lyase (Cth), and cysteinyl-tRNA synthetase 2 (Cars2). MPST deficiency was found in 1960s among rare hereditary mercaptolactate-cysteine disulfiduria patients. Mpst-knockout (KO) mice with enhanced liver Tst expression were recently generated as its model; however, the physiological roles/significances of Mpst remain largely unknown. Here we generated three independent germ lines of Mpst-KO mice by CRISPR/Cas9 technology, all of which maintained normal hepatic Tst expression/activity. Mpst/Cth-double knockout (DKO) mice were generated via crossbreeding with our previously generated Cth-KO mice. Mpst-KO mice were born at the expected frequency and developed normally like Cth-KO mice, but displayed increased urinary 3-mercaptolactate excretion and enhanced passive systemic anaphylactic responses when compared to wild-type or Cth-KO mice. Mpst/Cth-DKO mice were also born at the expected frequency and developed normally, but excreted slightly more 3-mercaptolactate in urine compared to Mpst-KO or Cth-KO mice. Our Mpst-KO, Cth-KO, and Mpst/Cth-DKO mice, unlike semi-lethal Cbs-KO mice and lethal Cars2-KO mice, are useful tools for analyzing the unknown physiological roles of endogenous H2S/RSS production.
Collapse
|
20
|
Akahoshi N, Yokoyama A, Nagata T, Miura A, Kamata S, Ishii I. Abnormal Amino Acid Profiles of Blood and Cerebrospinal Fluid from Cystathionine β-Synthase-Deficient Mice, an Animal Model of Homocystinuria. Biol Pharm Bull 2019; 42:1054-1057. [PMID: 31155583 DOI: 10.1248/bpb.b19-00127] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mental retardation is the most common feature among inborn errors of amino acid metabolism. Patients with homocystinuria/homocysteinemia caused by cystathionine β-synthase (CBS) deficiency suffer from thromboembolism and mental retardation from early ages; therefore, detection by newborn screening is performed. Furthermore, elevated levels of serum homocysteine during pregnancy are associated with the occurrence of neural tube defects (NTDs) in newborns. However, the causes of such central nervous system (CNS) defects are unknown. We found previously impaired learning abilities in Cbs-deficient (Cbs-/-) mice (but not NTD births). Here, we investigated the amino acid profiles of serum and cerebrospinal fluid (CSF) from Cbs-/- mice. Mice deficient in cystathionine γ-lyase (Cth), a downstream enzyme of CBS in transsulfuration, as well as wild-type mice, were analyzed as controls. Cbs-/- and Cth-/- mice were smaller than wild-type mice, and CSF yields in Cbs-/- mice were lower than the others. CSF amino acid levels were generally lower than those in serum, and compared with the dramatic amino acid level alterations in Cbs-/- mouse serum, alterations in CSF were less apparent. However, marked upregulation (versus wild-type) of aspartic acid/asparagine (Asp/Asn), glutamine (Gln), serine (Ser), threonine (Thr), phenylalanine (Phe), tyrosine (Tyr), methionine (Met), total homocysteine, and citrulline, and downregulation of lysine (Lys) were found in Cbs-/- mouse CSF. Because similar regulation of total homocysteine/citrulline/Lys was observed in the CSF of Cth-/- mice, which are free of CNS dysfunction, the reduced CSF volumes and the level changes of other amino acids could be relevant to Cbs-/--specific CNS defects.
Collapse
Affiliation(s)
| | - Akira Yokoyama
- Department of Health Chemistry, Showa Pharmaceutical University
| | - Tomoko Nagata
- Department of Biochemistry, Keio University Faculty of Pharmacy
| | - Asumi Miura
- Department of Biochemistry, Keio University Faculty of Pharmacy
| | - Shotaro Kamata
- Department of Health Chemistry, Showa Pharmaceutical University.,Department of Biochemistry, Keio University Faculty of Pharmacy
| | - Isao Ishii
- Department of Health Chemistry, Showa Pharmaceutical University.,Department of Biochemistry, Keio University Faculty of Pharmacy
| |
Collapse
|
21
|
Yang Q, He GW. Imbalance of Homocysteine and H 2S: Significance, Mechanisms, and Therapeutic Promise in Vascular Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:7629673. [PMID: 31885816 PMCID: PMC6893243 DOI: 10.1155/2019/7629673] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/16/2019] [Indexed: 12/28/2022]
Abstract
While the role of hyperhomocysteinemia in cardiovascular pathogenesis continuously draws attention, deficiency of hydrogen sulfide (H2S) has been growingly implicated in cardiovascular diseases. Generation of H2S is closely associated with the metabolism of homocysteine via key enzymes such as cystathionine β-synthase (CBS) and cystathionine γ-lyase (CSE). The level of homocysteine and H2S is regulated by each other. Metabolic switch in the activity of CBS and CSE may occur with a resultant operating preference change of these enzymes in homocysteine and H2S metabolism. This paper presented an overview regarding (1) linkage between the metabolism of homocysteine and H2S, (2) mutual regulation of homocysteine and H2S, (3) imbalance of homocysteine and H2S in cardiovascular disorders, (4) mechanisms underlying the protective effect of H2S against homocysteine-induced vascular injury, and (5) the current status of homocysteine-lowering and H2S-based therapies for cardiovascular disease. The metabolic imbalance of homocysteine and H2S renders H2S/homocysteine ratio a potentially reliable biomarker for cardiovascular disease and development of drugs or interventions targeting the interplay between homocysteine and H2S to maintain the endogenous balance of these two molecules may hold an even bigger promise for management of vascular disorders than targeting homocysteine or H2S alone.
Collapse
Affiliation(s)
- Qin Yang
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
| | - Guo-Wei He
- Center for Basic Medical Research & Department of Cardiovascular Surgery, TEDA International Cardiovascular Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China
- School of Pharmacy, Wannan Medical College, Wuhu, Anhui, China
- Department of Surgery, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
22
|
Akahoshi N, Handa H, Takemoto R, Kamata S, Yoshida M, Onaka T, Ishii I. Preeclampsia-Like Features and Partial Lactation Failure in Mice Lacking Cystathionine γ-Lyase-An Animal Model of Cystathioninuria. Int J Mol Sci 2019; 20:ijms20143507. [PMID: 31319489 PMCID: PMC6679037 DOI: 10.3390/ijms20143507] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Revised: 07/10/2019] [Accepted: 07/16/2019] [Indexed: 01/01/2023] Open
Abstract
Elevated plasma homocysteine levels are considered as a risk factor for cardiovascular diseases as well as preeclampsia—a pregnancy disorder characterized by hypertension and proteinuria. We previously generated mice lacking cystathionine γ-lyase (Cth) as cystathioninuria models and found them to be with cystathioninemia/homocysteinemia. We investigated whether Cth-deficient (Cth−/−) pregnant mice display any features of preeclampsia. Cth−/− females developed normally but showed mild hypertension (~10 mmHg systolic blood pressure elevation) in late pregnancy and mild proteinuria throughout development/pregnancy. Cth−/− dams had normal numbers of pups and exhibited normal maternal behavior except slightly lower breastfeeding activity. However, half of them could not raise their pups owing to defective lactation; they could produce/store the first milk in their mammary glands but not often provide milk to their pups after the first ejection. The serum oxytocin levels and oxytocin receptor expression in the mammary glands were comparable between wild-type and Cth−/− dams, but the contraction responses of mammary gland myoepithelial cells to oxytocin were significantly lower in Cth−/− dams. The contraction responses to oxytocin were lower in uteruses isolated from Cth−/− mice. Our results suggest that elevated homocysteine or other unknown factors in preeclampsia-like Cth−/− dams interfere with oxytocin that regulates milk ejection reflex.
Collapse
Affiliation(s)
- Noriyuki Akahoshi
- Department of Health Chemistry, Showa Pharmaceutical University, Tokyo 194-8543, Japan
| | - Hiroki Handa
- Department of Health Chemistry, Showa Pharmaceutical University, Tokyo 194-8543, Japan
| | - Rintaro Takemoto
- Department of Health Chemistry, Showa Pharmaceutical University, Tokyo 194-8543, Japan
| | - Shotaro Kamata
- Department of Health Chemistry, Showa Pharmaceutical University, Tokyo 194-8543, Japan
| | - Masahide Yoshida
- Department of Physiology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Tatsushi Onaka
- Department of Physiology, Jichi Medical University, Tochigi 329-0498, Japan
| | - Isao Ishii
- Department of Health Chemistry, Showa Pharmaceutical University, Tokyo 194-8543, Japan.
| |
Collapse
|
23
|
Kawano Y, Shiroyama M, Kanazawa K, Suzuki YA, Ohtsu I. Development of high-throughput quantitative analytical method for L-cysteine-containing dipeptides by LC-MS/MS toward its fermentative production. AMB Express 2019; 9:91. [PMID: 31227937 PMCID: PMC6588663 DOI: 10.1186/s13568-019-0817-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 06/15/2019] [Indexed: 11/23/2022] Open
Abstract
l-Cysteine (Cys) is metabolically fundamental sulfur compound and important components in various cellular factors. Interestingly, free-form Cys itself as a simple monomeric amino acid was recently shown to function in a novel antioxidative system (cysteine/cystine shuttle system) in Escherichia coli. However, as for Cys-containing dipeptides, the biological functions, effects, and even contents have still remained largely elusive. The potential functions should be a part of cellular redox system and important in basic and applied biology. For its progress, establishment of reliable quantitation method is the first. However, such accurate analysis is unexpectedly difficult even in Cys, because thiol compounds convert through disulfide-exchange and air oxidation during sample preparation. Addressing this problem, in this study, thiol molecules like Cys-containing dipeptides were derivatized by using monobromobimane (thiol-specific alkylating reagent) and detected as S-bimanyl derivatives by liquid chromatography coupled to tandem mass spectrometry (LC–MS/MS). Sample separation was processed with a C18 column (2.1 mm × 150 mm, 1.7 μm) and with water-acetonitrile gradient mobile phase containing 0.1% (v/v) formic acid at flow rate of 0.25 ml/min. The mass spectrometer was operated in the multiple reaction monitoring in positive/negative mode with electrospray ionization. The derivatization could indeed avoid the unfavorable reactions, namely, developed the method reflecting their correct contents on sampling. Furthermore, the method was successfully applied to monitoring Cys-containing dipeptides in E. coli Cys producer overexpressing bacD gene. This is the first report of the quantitative analysis of Cys-containing dipeptides, which should be useful for further study of fermentative production of Cys-containing dipeptides.
Collapse
|
24
|
Ni T, Gao F, Zhang J, Lin H, Luo H, Chi J, Guo H. Impaired autophagy mediates hyperhomocysteinemia-induced HA-VSMC phenotypic switching. J Mol Histol 2019; 50:305-314. [PMID: 31028566 DOI: 10.1007/s10735-019-09827-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Accepted: 04/22/2019] [Indexed: 01/07/2023]
Abstract
Hyperhomocysteinemia (HHcy) is a highly-related risk factor in vascular smooth muscle cell (VSMC) phenotypic modulation and atherosclerosis. Growing evidence indicated that autophagy is involved in pathological arterial changes. However, the risk mechanisms by which homocysteine and VSMC autophagy interact with cardiovascular disease are poorly understood. This study verified the homocysteine-responsive endoplasmic reticulum protein promotion of VSMC phenotypic switching, and the formation of atherosclerotic plaque in vitro. We found that impaired autophagy, as evidenced by decreased levels of MAP1LC3B II/MAP1LC3B I, has a vital role in HHcy-induced human aortic (HA)-VSMC phenotypic switching, with a decrease in contractile proteins (SM α-actin and calponin) and an increase in osteopontin. Knockdown of the essential autophagy gene Atg7 by small interfering RNA promoted HA-VSMC phenotypic switching, indicating that impaired autophagy induces phenotypic switching in these cells. HHcy co-treatment with rapamycin triggered autophagy, which alleviated HA-VSMC phenotypic switching. Finally, we found that Krüppel-like factor 4 (KLF4), a zinc-finger transcription factor for maintaining genomic stability by resisting oxidative stress and restoring autophagy, is closely involved in this process. HHcy clearly decreased KLF4 expression. KLF4-specific siRNA aggravated defective autophagy and phenotypic switching. Mechanistically, KLF4 regulated the HHcy-induced decrease in HA-VSMC autophagy via the m-TOR signaling pathway. In conclusion, these results demonstrated that the KLF4-dependent rapamycin signaling pathway is a novel mechanism underlying HA-VSMC phenotypic switching and is crucial for HHcy-induced HA-VSMCs with defective autophagy to accelerate early atherosclerosis.
Collapse
Affiliation(s)
- Tingjuan Ni
- Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, China
| | - Feidan Gao
- Zhejiang Chinese Medical University, Hangzhou, 310012, Zhejiang, China
| | - Jie Zhang
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hui Lin
- The First Clinical Medical College, Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Hangqi Luo
- Zhejiang University School of Medicine, Hangzhou, 310000, Zhejiang, China
| | - Jufang Chi
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, China.
| | - Hangyuan Guo
- Department of Cardiology, Shaoxing People's Hospital (Shaoxing Hospital Zhejiang University School of Medicine), No. 568 Zhongxing North Road, Shaoxing, 312000, Zhejiang, China.
| |
Collapse
|
25
|
Zhang L, Wang Y, Li Y, Li L, Xu S, Feng X, Liu S. Hydrogen Sulfide (H 2S)-Releasing Compounds: Therapeutic Potential in Cardiovascular Diseases. Front Pharmacol 2018; 9:1066. [PMID: 30298008 PMCID: PMC6160695 DOI: 10.3389/fphar.2018.01066] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 09/03/2018] [Indexed: 01/03/2023] Open
Abstract
Cardiovascular disease is the main cause of death worldwide, but its pathogenesis is not yet clear. Hydrogen sulfide (H2S) is considered to be the third most important endogenous gasotransmitter in the organism after carbon monoxide and nitric oxide. It can be synthesized in mammalian tissues and can freely cross the cell membrane and exert many biological effects in various systems including cardiovascular system. More and more recent studies have supported the protective effects of endogenous H2S and exogenous H2S-releasing compounds (such as NaHS, Na2S, and GYY4137) in cardiovascular diseases, such as cardiac hypertrophy, heart failure, ischemia/reperfusion injury, and atherosclerosis. Here, we provided an up-to-date overview of the mechanistic actions of H2S as well as the therapeutic potential of various classes of H2S donors in treating cardiovascular diseases.
Collapse
Affiliation(s)
- Lei Zhang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yanan Wang
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Yi Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Lingli Li
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Suowen Xu
- Aab Cardiovascular Research Institute, University of Rochester, Rochester, NY, United States
| | - Xiaojun Feng
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Sheng Liu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
26
|
Zhou J, Lv XH, Fan JJ, Dang LY, Dong K, Gao B, Song AQ, Wu WN. GYY4137 Promotes Mice Feeding Behavior via Arcuate Nucleus Sulfur-Sulfhydrylation and AMPK Activation. Front Pharmacol 2018; 9:966. [PMID: 30186182 PMCID: PMC6111581 DOI: 10.3389/fphar.2018.00966] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2018] [Accepted: 08/06/2018] [Indexed: 12/11/2022] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous gaseous molecule and plays important biological and neurochemical roles in many processes such as the neural activity and immunity. The arcuate nucleus (ARC) of hypothalamus is a control center for appetite and energy metabolism. AMPK is a gage kinase in the monitoring of energy status and regulation of energy metabolism, and it can be activated by H2S via CaMKKβ/AMPK pathway. But the role of H2S in ARC and appetite has not been reported. Here we studied the orexigenic effect of H2S and the mechanisms by means of GYY4137, a water soluble and slow-releasing donor of H2S, and protein sulfur-sulfhydrylation analysis. We demonstrated that GYY4137-derived H2S increased food intake of mice, augmented the production of neuropeptide Y (NPY), and elevated the protein sulfur-sulfhydrylation level and the activation of AMPK and CaMKKβ in ARC. Blocking sulfur-sulfhydrylation with DTT eliminated GYY4137-induced activation of AMPK and CaMKKβ. DTT and preventing AMPK activation in ARC with Compound C and Ara-A could both attenuate the orexigenic effect of GYY4137. These findings suggest that H2S enhances appetite through protein sulfur-sulfhydrylation and the activation of AMPK and NPY function in ARC.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Pharmacy, Xi'an Chest Hospital, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Xiao-Hui Lv
- Department of Pharmacy, Xi'an Chest Hospital, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Jun-Juan Fan
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Anhui Medical University, Hefei, China
| | - Li-Yun Dang
- Department of Pharmacy, Xi'an Chest Hospital, Shaanxi University of Chinese Medicine, Xi'an, China
| | - Kun Dong
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Anhui Medical University, Hefei, China
| | - Bo Gao
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Anhui Medical University, Hefei, China
| | - Ao-Qi Song
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Anhui Medical University, Hefei, China
| | - Wen-Ning Wu
- Department of Pharmacology, School of Basic Medical Sciences, Key Laboratory of Anti-inflammatory and Immunopharmacology, Anhui Medical University, Hefei, China
| |
Collapse
|
27
|
Kamata S, Yamamoto J, Ohtani H, Tosaka Y, Yoshikawa S, Akahoshi N, Ishii I. 2D DIGE proteomic analysis reveals fasting-induced protein remodeling through organ-specific transcription factor(s) in mice. FEBS Open Bio 2018; 8:1524-1543. [PMID: 30186752 PMCID: PMC6120221 DOI: 10.1002/2211-5463.12497] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/27/2018] [Accepted: 07/23/2018] [Indexed: 12/15/2022] Open
Abstract
Overnight fasting is a routine procedure before surgery in clinical settings. Intermittent fasting is the most common diet/fitness trend implemented for weight loss and the treatment of lifestyle‐related diseases. In either setting, the effects not directly related to parameters of interest, either beneficial or harmful, are often ignored. We previously demonstrated differential activation of cellular adaptive responses in 13 atrophied/nonatrophied organs of fasted mice by quantitative PCR analysis of gene expression. Here, we investigated 2‐day fasting‐induced protein remodeling in six major mouse organs (liver, kidney, thymus, spleen, brain, and testis) using two‐dimensional difference gel electrophoresis (2D DIGE) proteomics as an alternative means to examine systemic adaptive responses. Quantitative analysis of protein expression followed by protein identification using matrix‐assisted laser desorption ionization–time‐of‐flight mass spectrometry (MALDI‐TOFMS) revealed that the expression levels of 72, 26, and 14 proteins were significantly up‐ or downregulated in the highly atrophied liver, thymus, and spleen, respectively, and the expression levels of 32 proteins were up‐ or downregulated in the mildly atrophied kidney. Conversely, there were no significant protein expression changes in the nonatrophied organs, brain and testis. Upstream regulator analysis highlighted transcriptional regulation by peroxisome proliferator‐activated receptor alpha (PPARα) in the liver and kidney and by tumor protein/suppressor p53 (TP53) in the thymus, spleen, and liver. These results imply of the existence of both common and distinct adaptive responses between major mouse organs, which involve transcriptional regulation of specific protein expression upon short‐term fasting. Our data may be valuable in understanding systemic transcriptional regulation upon fasting in experimental animals.
Collapse
Affiliation(s)
- Shotaro Kamata
- Laboratory of Health Chemistry Showa Pharmaceutical University Tokyo Japan.,Laboratory of Biochemistry Keio University School of Pharmaceutical Sciences Tokyo Japan
| | - Junya Yamamoto
- Laboratory of Biochemistry Keio University School of Pharmaceutical Sciences Tokyo Japan
| | - Haruka Ohtani
- Laboratory of Biochemistry Keio University School of Pharmaceutical Sciences Tokyo Japan
| | - Yuka Tosaka
- Laboratory of Biochemistry Keio University School of Pharmaceutical Sciences Tokyo Japan
| | - Sayumi Yoshikawa
- Laboratory of Biochemistry Keio University School of Pharmaceutical Sciences Tokyo Japan
| | - Noriyuki Akahoshi
- Laboratory of Health Chemistry Showa Pharmaceutical University Tokyo Japan
| | - Isao Ishii
- Laboratory of Health Chemistry Showa Pharmaceutical University Tokyo Japan.,Laboratory of Biochemistry Keio University School of Pharmaceutical Sciences Tokyo Japan
| |
Collapse
|
28
|
Kawano Y, Suzuki K, Ohtsu I. Current understanding of sulfur assimilation metabolism to biosynthesize L-cysteine and recent progress of its fermentative overproduction in microorganisms. Appl Microbiol Biotechnol 2018; 102:8203-8211. [PMID: 30046857 DOI: 10.1007/s00253-018-9246-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/01/2022]
Abstract
To all organisms, sulfur is an essential and important element. The assimilation of inorganic sulfur molecules such as sulfate and thiosulfate into organic sulfur compounds such as L-cysteine and L-methionine (essential amino acid for human) is largely contributed by microorganisms. Of these, special attention is given to thiosulfate (S2O32-) assimilation, because thiosulfate relative to often utilized sulfate (SO42-) as a sulfur source is proposed to be more advantageous in microbial growth and biotechnological applications like L-cysteine fermentative overproduction toward industrial manufacturing. In Escherichia coli as well as other many bacteria, the thiosulfate assimilation pathway is known to depend on O-acetyl-L-serine sulfhydrylase B. Recently, another yet-unidentified CysM-independent thiosulfate pathway was found in E. coli. This pathway is expected to consist of the initial part of the thiosulfate to sulfite (SO32-) conversion, and the latter part might be shared with the final part of the known sulfate assimilation pathway [sulfite → sulfide (S2-) → L-cysteine]. The catalysis of thiosulfate to sulfite is at least partly mediated by thiosulfate sulfurtransferase (GlpE). In this mini-review, we introduce updated comprehensive information about sulfur assimilation in microorganisms, including this topic. Also, we introduce recent advances of the application study about L-cysteine overproduction, including the GlpE overexpression.
Collapse
Affiliation(s)
- Yusuke Kawano
- Innovation Medical Research Institute, University of Tsukuba, Tsukuba, Japan
| | - Kengo Suzuki
- Department of Research and Development, Euglena Co., Ltd., Minato-ku, Tokyo, Japan
| | - Iwao Ohtsu
- Innovation Medical Research Institute, University of Tsukuba, Tsukuba, Japan.
| |
Collapse
|
29
|
Fujii S, Sawa T, Motohashi H, Akaike T. Persulfide synthases that are functionally coupled with translation mediate sulfur respiration in mammalian cells. Br J Pharmacol 2018; 176:607-615. [PMID: 29748969 DOI: 10.1111/bph.14356] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 04/16/2018] [Accepted: 04/26/2018] [Indexed: 12/13/2022] Open
Abstract
Cysteine persulfide and polysulfide are produced in cells and exist in abundance in both low MW and protein fractions. However, the mechanism of regulation of the formation of cellular cysteine polysulfides and the physiological functions of cysteine persulfides/polysulfides produced in cells are not fully understood. We recently demonstrated that cysteinyl-tRNA synthetase (CARS) is a novel cysteine persulfide synthase. CARS is involved in protein polysulfidation that is coupled with translation. In particular, mitochondria function in biogenesis and bioenergetics is also supported and up-regulated by cysteine persulfide derived from mitochondrial CARS (also known as CARS2). Here, we provide an overview of recent advances in reactive persulfide research and our understanding of the mechanisms underlying the formation and the physiological roles of reactive persufides, with a primary focus on the formation of cysteine persulfide by CARS and the most fundamental mitochondrial bioenergetics mediated by persulfides, that is, sulfur respiration. LINKED ARTICLES: This article is part of a themed section on Chemical Biology of Reactive Sulfur Species. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.4/issuetoc.
Collapse
Affiliation(s)
- Shigemoto Fujii
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hozumi Motohashi
- Department of Gene Expression Regulation, IDAC, Tohoku University, Sendai, Japan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
30
|
Gold-nanofève surface-enhanced Raman spectroscopy visualizes hypotaurine as a robust anti-oxidant consumed in cancer survival. Nat Commun 2018; 9:1561. [PMID: 29674746 PMCID: PMC5908798 DOI: 10.1038/s41467-018-03899-1] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2017] [Accepted: 03/20/2018] [Indexed: 01/24/2023] Open
Abstract
Gold deposition with diagonal angle towards boehmite-based nanostructure creates random arrays of horse-bean-shaped nanostructures named gold-nanofève (GNF). GNF generates many electromagnetic hotspots as surface-enhanced Raman spectroscopy (SERS) excitation sources, and enables large-area visualization of molecular vibration fingerprints of metabolites in human cancer xenografts in livers of immunodeficient mice with sufficient sensitivity and uniformity. Differential screening of GNF-SERS signals in tumours and those in parenchyma demarcated tumour boundaries in liver tissues. Furthermore, GNF-SERS combined with quantum chemical calculation identified cysteine-derived glutathione and hypotaurine (HT) as tumour-dominant and parenchyma-dominant metabolites, respectively. CD44 knockdown in cancer diminished glutathione, but not HT in tumours. Mechanisms whereby tumours sustained HT under CD44-knockdown conditions include upregulation of PHGDH, PSAT1 and PSPH that drove glycolysis-dependent activation of serine/glycine-cleavage systems to provide one-methyl group for HT synthesis. HT was rapidly converted into taurine in cancer cells, suggesting that HT is a robust anti-oxidant for their survival under glutathione-suppressed conditions. Surface-enhanced Raman spectroscopy (SERS) visualizes fingerprints of intermolecular vibrations of many metabolites. Here the authors report a SERS imaging technique that enables the visualization of metabolites distribution and automated extraction of tumour boundaries in frozen tissues.
Collapse
|
31
|
Cysteinyl-tRNA synthetase governs cysteine polysulfidation and mitochondrial bioenergetics. Nat Commun 2017; 8:1177. [PMID: 29079736 PMCID: PMC5660078 DOI: 10.1038/s41467-017-01311-y] [Citation(s) in RCA: 394] [Impact Index Per Article: 49.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 08/29/2017] [Indexed: 12/30/2022] Open
Abstract
Cysteine hydropersulfide (CysSSH) occurs in abundant quantities in various organisms, yet little is known about its biosynthesis and physiological functions. Extensive persulfide formation is apparent in cysteine-containing proteins in Escherichia coli and mammalian cells and is believed to result from post-translational processes involving hydrogen sulfide-related chemistry. Here we demonstrate effective CysSSH synthesis from the substrate l-cysteine, a reaction catalyzed by prokaryotic and mammalian cysteinyl-tRNA synthetases (CARSs). Targeted disruption of the genes encoding mitochondrial CARSs in mice and human cells shows that CARSs have a crucial role in endogenous CysSSH production and suggests that these enzymes serve as the principal cysteine persulfide synthases in vivo. CARSs also catalyze co-translational cysteine polysulfidation and are involved in the regulation of mitochondrial biogenesis and bioenergetics. Investigating CARS-dependent persulfide production may thus clarify aberrant redox signaling in physiological and pathophysiological conditions, and suggest therapeutic targets based on oxidative stress and mitochondrial dysfunction. Cysteine hydropersulfides (CysSSH) are believed to have a cellular redox protective role. Here the authors show that these species can be produced from L-cysteine by cysteinyl-tRNA synthetases and that these enzymes are also involved in mitochondrial biogenesis and bioenergetics regulation.
Collapse
|
32
|
Improved fermentative l-cysteine overproduction by enhancing a newly identified thiosulfate assimilation pathway in Escherichia coli. Appl Microbiol Biotechnol 2017; 101:6879-6889. [DOI: 10.1007/s00253-017-8420-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/09/2017] [Accepted: 07/02/2017] [Indexed: 10/19/2022]
|
33
|
Hine C, Kim HJ, Zhu Y, Harputlugil E, Longchamp A, Matos MS, Ramadoss P, Bauerle K, Brace L, Asara JM, Ozaki CK, Cheng SY, Singha S, Ahn KH, Kimmelman A, Fisher FM, Pissios P, Withers DJ, Selman C, Wang R, Yen K, Longo VD, Cohen P, Bartke A, Kopchick JJ, Miller R, Hollenberg AN, Mitchell JR. Hypothalamic-Pituitary Axis Regulates Hydrogen Sulfide Production. Cell Metab 2017; 25:1320-1333.e5. [PMID: 28591635 PMCID: PMC5722247 DOI: 10.1016/j.cmet.2017.05.003] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Revised: 01/20/2017] [Accepted: 05/11/2017] [Indexed: 01/27/2023]
Abstract
Decreased growth hormone (GH) and thyroid hormone (TH) signaling are associated with longevity and metabolic fitness. The mechanisms underlying these benefits are poorly understood, but may overlap with those of dietary restriction (DR), which imparts similar benefits. Recently we discovered that hydrogen sulfide (H2S) is increased upon DR and plays an essential role in mediating DR benefits across evolutionary boundaries. Here we found increased hepatic H2S production in long-lived mouse strains of reduced GH and/or TH action, and in a cell-autonomous manner upon serum withdrawal in vitro. Negative regulation of hepatic H2S production by GH and TH was additive and occurred via distinct mechanisms, namely direct transcriptional repression of the H2S-producing enzyme cystathionine γ-lyase (CGL) by TH, and substrate-level control of H2S production by GH. Mice lacking CGL failed to downregulate systemic T4 metabolism and circulating IGF-1, revealing an essential role for H2S in the regulation of key longevity-associated hormones.
Collapse
Affiliation(s)
- Christopher Hine
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Hyo-Jeong Kim
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Yan Zhu
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Eylul Harputlugil
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Alban Longchamp
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Surgery, Heart and Vascular Center Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Marina Souza Matos
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Preeti Ramadoss
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Kevin Bauerle
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lear Brace
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - John M Asara
- Division of Signal Transduction, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - C Keith Ozaki
- Department of Surgery, Heart and Vascular Center Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Sheue-Yann Cheng
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Subhankar Singha
- Department of Chemistry, Center for Electro-Photo Behaviors in Advanced Molecular Systems, POSTECH, 77 Cheongam-Ro, Nam-Gu, Pohang 790-784, Republic of Korea
| | - Kyo Han Ahn
- Department of Chemistry, Center for Electro-Photo Behaviors in Advanced Molecular Systems, POSTECH, 77 Cheongam-Ro, Nam-Gu, Pohang 790-784, Republic of Korea
| | - Alec Kimmelman
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Ffolliott M Fisher
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Pavlos Pissios
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Dominic J Withers
- Medical Research Council Clinical Science Centre, Imperial College, London W12 0NN, UK
| | - Colin Selman
- Glasgow Ageing Research Network, Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK
| | - Rui Wang
- Department of Biology, Lakehead University, Thunder Bay, ON P7B 5E1, Canada
| | - Kelvin Yen
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Valter D Longo
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Pinchas Cohen
- Department of Biological Sciences, Longevity Institute, School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA
| | - Andrzej Bartke
- Department of Internal Medicine, Southern Illinois University School of Medicine, Springfield, IL 62794, USA
| | - John J Kopchick
- Edison Biotechnology Institute, Heritage College of Osteopathic Medicine, Ohio University, Athens, OH 45701, USA
| | - Richard Miller
- Department of Pathology & Geriatrics Center, University of Michigan, Ann Arbor, MI 48109, USA
| | - Anthony N Hollenberg
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | - James R Mitchell
- Department of Genetics and Complex Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
34
|
Zhang T, Wei W, Dirsch O, Krüger T, Kan C, Xie C, Kniemeyer O, Fang H, Settmacher U, Dahmen U. Identification of Proteins Interacting with Cytoplasmic High-Mobility Group Box 1 during the Hepatocellular Response to Ischemia Reperfusion Injury. Int J Mol Sci 2017; 18:ijms18010167. [PMID: 28275217 PMCID: PMC5297800 DOI: 10.3390/ijms18010167] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/03/2017] [Accepted: 01/09/2017] [Indexed: 01/22/2023] Open
Abstract
Ischemia/reperfusion injury (IRI) occurs inevitably in liver transplantations and frequently during major resections, and can lead to liver dysfunction as well as systemic disorders. High-mobility group box 1 (HMGB1) plays a pathogenic role in hepatic IRI. In the normal liver, HMGB1 is located in the nucleus of hepatocytes; after ischemia reperfusion, it translocates to the cytoplasm and it is further released to the extracellular space. Unlike the well-explored functions of nuclear and extracellular HMGB1, the role of cytoplasmic HMGB1 in hepatic IRI remains elusive. We hypothesized that cytoplasmic HMGB1 interacts with binding proteins involved in the hepatocellular response to IRI. In this study, binding proteins of cytoplasmic HMGB1 during hepatic IRI were identified. Liver tissues from rats with warm ischemia reperfusion (WI/R) injury and from normal rats were subjected to cytoplasmic protein extraction. Co-immunoprecipitation using these protein extracts was performed to enrich HMGB1-protein complexes. To separate and identify the immunoprecipitated proteins in eluates, 2-dimensional electrophoresis and subsequent mass spectrometry detection were performed. Two of the identified proteins were verified using Western blotting: betaine–homocysteine S-methyltransferase 1 (BHMT) and cystathionine γ-lyase (CTH). Therefore, our results revealed the binding of HMGB1 to BHMT and CTH in cytoplasm during hepatic WI/R. This finding may help to better understand the cellular response to IRI in the liver and to identify novel molecular targets for reducing ischemic injury.
Collapse
Affiliation(s)
- Tianjiao Zhang
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| | - Weiwei Wei
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| | - Olaf Dirsch
- Institute of Pathology, Klinikum Chemnitz gGmbH, 09116 Chemnitz, Germany.
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 07745 Jena, Germany.
| | - Chunyi Kan
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
- Department of Obstetrics and Gynecology, Wuhan Central Hospital, Wuhan 430014, China.
| | - Chichi Xie
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, 07745 Jena, Germany.
| | - Haoshu Fang
- Department of Pathophysiology, Anhui Medical University, Hefei 230032, China.
| | - Utz Settmacher
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General, Visceral and Vascular Surgery, Jena University Hospital, 07747 Jena, Germany.
| |
Collapse
|
35
|
Kabe Y, Yamamoto T, Kajimura M, Sugiura Y, Koike I, Ohmura M, Nakamura T, Tokumoto Y, Tsugawa H, Handa H, Kobayashi T, Suematsu M. Cystathionine β-synthase and PGRMC1 as CO sensors. Free Radic Biol Med 2016; 99:333-344. [PMID: 27565814 DOI: 10.1016/j.freeradbiomed.2016.08.025] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2016] [Revised: 08/21/2016] [Accepted: 08/23/2016] [Indexed: 11/30/2022]
Abstract
Heme oxygenase (HO) is a mono-oxygenase utilizing heme and molecular oxygen (O2) as substrates to generate biliverdin-IXα and carbon monoxide (CO). HO-1 is inducible under stress conditions, while HO-2 is constitutive. A balance between heme and CO was shown to regulate cell death and survival in many experimental models. However, direct molecular targets to which CO binds to regulate cellular functions remained to be fully examined. We have revealed novel roles of CO-responsive proteins, cystathionine β-synthase (CBS) and progesterone receptor membrane component 1 (PGRMC1), in regulating cellular functions. CBS possesses a prosthetic heme that allows CO binding to inhibit the enzyme activity and to regulate H2S generation and/or protein arginine methylation. On the other hand, in response to heme accumulation in cells, PGRMC1 forms a stable dimer through stacking interactions of two protruding heme molecules. Heme-mediated PGRMC1 dimerization is necessary to interact with EGF receptor and cytochromes P450 that determine cell proliferation and xenobiotic metabolism. Furthermore, CO interferes with PGRMC1 dimerization by dissociating the heme stacking, and thus results in modulation of cell responses. This article reviews the intriguing functions of these two proteins in response to inducible and constitutive levels of CO with their pathophysiological implications.
Collapse
Affiliation(s)
- Yasuaki Kabe
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo 160-8582, Japan
| | - Takehiro Yamamoto
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Mayumi Kajimura
- Department of Biology, Keio University School of Medicine, Yokohama 223-8521, Japan
| | - Yuki Sugiura
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Ikko Koike
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Mitsuyo Ohmura
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo 160-8582, Japan
| | - Takashi Nakamura
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan
| | - Yasuhito Tokumoto
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan; Admission Center, Saitama Medical University, Moroyama 350-0495, Japan
| | - Hitoshi Tsugawa
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan; Japan Agency for Medical Research and Development, Core Research for Evolutional Science and Technology (AMED-CREST), Tokyo 160-8582, Japan
| | - Hiroshi Handa
- Department of Nanoparticle Translational Research, Tokyo Medical University, Tokyo 160-8402, Japan
| | - Takuya Kobayashi
- Department of Medical Chemistry and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Makoto Suematsu
- Department of Biochemistry, Keio University School of Medicine, Tokyo 160-8582, Japan.
| |
Collapse
|
36
|
Abstract
Hydrogen sulfide (H(2)S) is a gasomediator synthesized from L- and D-cysteine in various tissues. It is involved in a number of physiological and pathological processes. H(2)S exhibits antiatherosclerotic, vasodilator, and proangiogenic properties, and protects the kidney and heart from damage following ischemia/reperfusion injury. H(2)S donors may be natural or synthetic, and may be used for the safe treatment of a wide range of diseases. This review article summarizes the current state of knowledge of the therapeutic function of H(2)S.
Collapse
Affiliation(s)
- Beata Olas
- Department of General Biochemistry, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland.
| |
Collapse
|
37
|
Yamamoto J, Kamata S, Miura A, Nagata T, Kainuma R, Ishii I. Differential adaptive responses to 1- or 2-day fasting in various mouse tissues revealed by quantitative PCR analysis. FEBS Open Bio 2015; 5:357-68. [PMID: 25973363 PMCID: PMC4420774 DOI: 10.1016/j.fob.2015.04.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Revised: 04/09/2015] [Accepted: 04/21/2015] [Indexed: 01/25/2023] Open
Abstract
Adaptive cellular responses to 1- or 2-day fasting differ among tissues. Ubiquitin–proteasome and autophagy–lysosome pathways are activated in thymus/lung/heart/muscle. The amino acid response is activated mainly in thymus. An Nrf2-mediated antioxidant system is activated in thymus, heart, and kidney. Expression of amino acid transporter genes is activated in a tissue-specific manner.
Dietary or caloric restriction confers various clinical benefits. Short-term fasting of mice is a common experimental procedure that may involve systemic metabolic remodeling, which may significantly affect experimental outputs. This study evaluated adaptive cellular responses after 1- or 2-day fasting in 13 mouse tissues by quantitative PCR using 15 marker primer sets for the activation of ubiquitin–proteasome (Atrogin-1 and MuRF1), autophagy–lysosome (LC3b, p62 and Lamp2), amino acid response (Asns, Trib3, Herpud1, xCT, and Chop), Nrf2-mediated antioxidant (HO-1 and Gsta1), and amino acid transport (Slc38a2, Slc7a5, and Slc7a1) systems. Differential activation profiles obtained in seven highly (thymus, liver, spleen, and small intestine) or mildly (stomach, kidney, and colon) atrophied tissues as well as in six non-atrophied tissues (brain, eye, lung, heart, skeletal muscle, and testis) suggested tissue-specific active metabolic remodeling.
Collapse
Affiliation(s)
- Junya Yamamoto
- Department of Biochemistry, Keio University Graduate School of Pharmaceutical Sciences, Japan
| | - Shotaro Kamata
- Department of Biochemistry, Keio University Graduate School of Pharmaceutical Sciences, Japan
| | - Asumi Miura
- Department of Biochemistry, Keio University Graduate School of Pharmaceutical Sciences, Japan
| | - Tomoko Nagata
- Department of Biochemistry, Keio University Graduate School of Pharmaceutical Sciences, Japan
| | - Ryo Kainuma
- Department of Biochemistry, Keio University Graduate School of Pharmaceutical Sciences, Japan
| | - Isao Ishii
- Department of Biochemistry, Keio University Graduate School of Pharmaceutical Sciences, Japan
| |
Collapse
|