1
|
Wang Y, Zheng Y, Liang X, Chang Y, Liu Y, Cheng X, Zhang M, Gao W, Li T. α-Lipoic acid alleviate myocardial infarction by suppressing age-independent macrophage senescence. Sci Rep 2025; 15:11996. [PMID: 40199978 PMCID: PMC11978910 DOI: 10.1038/s41598-025-92328-7] [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: 07/08/2024] [Accepted: 02/26/2025] [Indexed: 04/10/2025] Open
Abstract
Myocardial infarction (MI) has high morbidity and mortality, and the macrophage senescence-associated secretory phenotype (SASP) plays a central role in M1 healing. α-Lipoic acid (ALA) alleviates MI by regulating the function of macrophages, although the relationship between ALA and macrophage senescence remains unclear. To investigate macrophage SASP in MI, we performed single-cell RNA sequencing (scRNA-seq) on the GEO GSE163465 dataset, along with qPCR and western blot analyses to assess SASP expression in macrophages subjected to hypoxia and ALA treatment. Immunofluorescence was used to detect SASP distribution. Coculture and animal experiments were performed to assess the therapeutic effects of ALA on macrophage senescence and cardiomyocyte ischemic injury. scRNA-seq revealed an age-independent senescent propensity of macrophages in MI. Increased expression of H2A.X, CCL7, IL1β, and CDKN1A, along with decreased SOD2 expression, confirmed that macrophage SASP occurred after hypoxia, with oxidative stress and energy metabolism involved in the process. ALA inhibited the degradation of SIRT1 and promoted the Nrf2 nuclear translocation, alleviating macrophage senescence and myocardial ischemic injury. Age-independent macrophage SASP occurred during MI. Macrophage SASP was induced by ROS and mitochondrial dysfunction. ALA alleviated SASP by decreasing ROS generation and autophagy flux while increasing SIRT1 levels, and Nrf2 nuclear translocation. ALA ameliorated MI injury.
Collapse
Affiliation(s)
- Yuchao Wang
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
- Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Tianjin ECMO Treatment and Training Base, Tianjin, 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yue Zheng
- School of Medicine, Nankai University, Tianjin, 300071, China
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
- Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Tianjin ECMO Treatment and Training Base, Tianjin, 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xiaoyu Liang
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
- Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin, 300170, China
- The Third Central, Clinical College of Tianjin Medical University, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Tianjin ECMO Treatment and Training Base, Tianjin, 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yun Chang
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
- Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Tianjin ECMO Treatment and Training Base, Tianjin, 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Yanwu Liu
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
- Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin, 300170, China
- The Third Central, Clinical College of Tianjin Medical University, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Tianjin ECMO Treatment and Training Base, Tianjin, 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Xian Cheng
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
- Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin, 300170, China
- The Third Central, Clinical College of Tianjin Medical University, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Tianjin ECMO Treatment and Training Base, Tianjin, 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Meng Zhang
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
- Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin, 300170, China
- The Third Central, Clinical College of Tianjin Medical University, Tianjin, 300170, China
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China
- Tianjin ECMO Treatment and Training Base, Tianjin, 300170, China
- Artificial Cell Engineering Technology Research Center, Tianjin, China
| | - Wenqing Gao
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China.
- Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin, 300170, China.
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China.
- Tianjin ECMO Treatment and Training Base, Tianjin, 300170, China.
- Artificial Cell Engineering Technology Research Center, Tianjin, China.
| | - Tong Li
- School of Medicine, Nankai University, Tianjin, 300071, China.
- Department of Heart Center, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China.
- Nankai University Affiliated Third Center Hospital, No. 83, Jintang Road, Hedong District, Tianjin, 300170, China.
- The Third Central, Clinical College of Tianjin Medical University, Tianjin, 300170, China.
- Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China.
- Tianjin ECMO Treatment and Training Base, Tianjin, 300170, China.
- Artificial Cell Engineering Technology Research Center, Tianjin, China.
| |
Collapse
|
2
|
Xu Y, Sui Y, Jiang R, Wang X, Suda M, Niimi M, Mao Z, Zhang Z, Zhang SL, Fan J, Yao J. Sulfhydrated albumin transmits H 2S signaling and ameliorates DOX-induced multiorgan injuries. Redox Biol 2025; 83:103631. [PMID: 40228337 DOI: 10.1016/j.redox.2025.103631] [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: 03/10/2025] [Revised: 04/01/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
Hydrogen sulfide (H2S) is a vital signaling molecule involved in various physiological processes; however, the mechanisms underlying its systemic signaling remain poorly understood. We hypothesized that albumin, the predominant plasma protein and a vital sulfhydryl carrier, mediated systemic H2S signaling, which could potentially treat H2S-deficient diseases. This study aimed to investigate this hypothesis. Our results showed the presence of sulfhydrated proteins in normal mouse serum, with albumin being particularly enriched. The level of sulfhydration was influenced by H2S availability and the redox environment. In vitro incubation of albumin with NaHS resulted in an increased number of sulfhydrated groups. Under reductive conditions, this sulfhydrated albumin (-SSH-Alb) released substantial amounts of H2S. When -SSH-Alb was added to cultured endothelial cells, it activated the cAMP signaling pathway, upregulated cystathionine γ-lyase (CSE) expression, and enhanced intracellular H2S levels. In an in vitro inflammatory model involving macrophages and endothelial cells, -SSH-Alb inhibited macrophage adhesion, reduced LPS-induced expression of adhesion molecules, and suppressed cytokine production and inflammasome activation. These effects correlated with improved cellular redox status. Furthermore, in vivo administration of -SSH-Alb protected mice from doxorubicin (DOX)-induced cardiotoxicity and intestinal damage. It improved mouse mortality, and alleviated ferroptotic cardiac injury and gut barrier dysfunction. These therapeutic benefits were associated with rebalanced local and systemic redox status. In summary, our study reveals that -SSH-Alb reserves, transmits, and amplifies H2S signals and exhibits significant anti-inflammatory and antioxidant properties. This characteristic of -SSH-Alb holds promise for preventing and treating a wide range of diseases.
Collapse
Affiliation(s)
- Yijun Xu
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Yang Sui
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Rui Jiang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Xin Wang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Mika Suda
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Manabu Niimi
- Division of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Zhimin Mao
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Zhen Zhang
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan
| | - Shao-Ling Zhang
- Department of Medicine, Université de Montréal, Centre de recherche du Centre hospitalier de l'Université de Montréal (CRCHUM), 900 Saint Denis Street, H2 X 0A9, Montréal, QC, Canada
| | - Jianglin Fan
- Division of Molecular Pathology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan; Guangdong Province Key Laboratory, Southern China Institute of Large Animal Models for Biomedicine, School of Pharmacy and Food Engineering, Wuyi University, Jiangmen, China.
| | - Jian Yao
- Division of Molecular Signaling, Department of the Advanced Biomedical Research, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, 409-3898, Japan.
| |
Collapse
|
3
|
De Brasi-Velasco S, Aroca A, Romero LC, Gotor C, Sevilla F, Jiménez A. New role for thioredoxins in plants: Implication of TRXo1 in protein depersulfidation. Redox Biol 2025; 82:103627. [PMID: 40220625 DOI: 10.1016/j.redox.2025.103627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 03/26/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025] Open
Abstract
Persulfidation, a posttranslational modification of cysteines to persulfides, is the best characterized molecular mechanism of H2S signaling. This study is focused on new functions for thioredoxins (TRXs) in plants beyond those of thiol disulfide (S-S) exchange, including the regulation of protein persulfidation as it has been described in animal systems. To elucidate the impact of TRXo1 deficiency on the protein persulfidation pattern in plants of Arabidopsis thaliana L. wild type (WT) and two Attrxo1 T-DNA insertion mutants grown under non stress conditions, a quantitative proteomic approach was performed. The proteomic analysis revealed a higher number of proteins that were more persulfidated in the mutants compared to WT plants, suggesting a role for TRXo1 in protein depersulfidation. Interestingly, most of the differentially persulfidated proteins were located in the chloroplast, implying a coordination between chloroplast H2S-dependent persulfidation and mitochondrial TRXo1 depersulfidation. Among the differentially persulfidated proteins located in mitochondria, the antioxidant enzymes sAPX, DHAR1 and MDAR6 were selected for further studies. The effect of H2S-dependent persulfidation on their enzymatic activities and its reversibility by the NADPH/thioredoxin reductase (NTRB)/TRXo1 system was analyzed, as well as their persulfidation levels were quantified. Sulfide treatment brought about increases in the activity levels of the enzymes, that match with a raise on the persulfidation levels. Interestingly, both activations declining after treatment with the thioredoxin system, indicate the regulation of their persulfidation by TRXo1. These results point to a positive effect of persulfidation on the enzymatic activities and also to a new depersulfidase activity for TRXo1. All together these results give a new insight of the mechanism of elimination of -SSH groups in plants exerted by TRXo1, and the involvement of a redox regulation on the protein persulfidation.
Collapse
Affiliation(s)
| | - Angeles Aroca
- Instituto de Bioquímica Vegetal y Fotosíntesis (CSIC-US), Sevilla, Spain
| | - Luis C Romero
- Instituto de Bioquímica Vegetal y Fotosíntesis (CSIC-US), Sevilla, Spain
| | - Cecilia Gotor
- Instituto de Bioquímica Vegetal y Fotosíntesis (CSIC-US), Sevilla, Spain.
| | - Francisca Sevilla
- Centro de Edafología y Biología Aplicada del Segura-CSIC, Murcia, Spain
| | - Ana Jiménez
- Centro de Edafología y Biología Aplicada del Segura-CSIC, Murcia, Spain.
| |
Collapse
|
4
|
Zhang T, Toyomoto T, Sawa T, Akaike T, Matsunaga T. Supersulfides: A Promising Therapeutic Approach for Autoinflammatory Diseases. Microbiol Immunol 2025; 69:191-202. [PMID: 39956868 PMCID: PMC11973847 DOI: 10.1111/1348-0421.13205] [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: 01/23/2025] [Revised: 02/04/2025] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
Supersulfides are molecular species characterized by catenated sulfur moieties, including low-molecular-weight and protein-bound supersulfides. Emerging evidence suggests that these molecules, abundantly present in diverse organisms, play essential roles far beyond their chemical properties, such as functions in energy metabolism, protein stabilization, and antiviral defense. Recent studies highlight their regulatory effects on pattern-recognition receptors (PRRs) and associated signaling pathways-such as nucleotide oligomerization domain-like receptor signaling, toll-like receptor signaling, and type I interferon receptor signaling-critical for innate immunity and inflammatory responses. Dysregulation of these pathways is implicated in a heterogeneous group of autoinflammatory diseases, including inflammasomopathies, relopathies, and type I interferonopathies, respectively. Notably, both endogenous and synthetic supersulfide donors have recently shown promising inhibitory effects on PRR signaling, offering their potential as targeted therapies for managing autoinflammatory conditions. This review summarizes the fundamental biology of supersulfides and typical autoinflammatory diseases, focusing on their roles in innate immune and inflammatory responses, while exploring their therapeutic potential in these diseases.
Collapse
Grants
- This work was supported by JST CREST Grant Number JPMJCR2024 (20348438 to T.A.), Grant-in-Aid for Scientific Research on Innovative Areas(A) "Sulfur biology" (21H05263 to T.A., 21H05267 to T.S., and 21H05258 to T.A. and T.S), International Leading Research (23K20040 to T.A.), Scientific Research (S) (24H00063 to T.A.), Challenge Research (Exploratory) (23K17979 to T.S.), Scientific Research (B) (22K06893 to T.M.), from the Ministry of Education, Culture, Sports, Science and Technology of Japan, and Japan Agency for Medical Research and Development (AMED) to T. Akaike (JP21zf0127001), and AMED CREST Grant Number 23gm161001h001 to T.S.
Collapse
Affiliation(s)
- Tianli Zhang
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious DiseasesAkita UniversityAkitaJapan
| | - Touya Toyomoto
- Department of Microbiology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Tomohiro Sawa
- Department of Microbiology, Graduate School of Medical SciencesKumamoto UniversityKumamotoJapan
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular ToxicologyTohoku University Graduate School of MedicineSendaiJapan
| | - Tetsuro Matsunaga
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious DiseasesAkita UniversityAkitaJapan
| |
Collapse
|
5
|
Feng H, Tian C, Jiang W, Sun Z, Li Y, Han B, Chen L, Wang D, Xiang H, Zhu J, Song W, Li J, Cai Y, Wang S, Li Y. Hydrogen sulfide sustains mitochondria functions via targeting mitochondria fission regulator 1 like protein to restore human cytotrophoblast invasion and migration. Int J Biol Macromol 2025; 299:140240. [PMID: 39854860 DOI: 10.1016/j.ijbiomac.2025.140240] [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: 11/26/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 01/27/2025]
Abstract
Hydrogen sulfide (H2S) is bioactive in mammals. Reduced H2S was observe in pregnancy complications, pre-eclampsia (PE). Our previous data demonstrated that low dose of H2S enhanced cytotrophoblast (CTB) invasion and migration via mitochondria dynamics without knowing the mechanisms. This study was designed to explore the functional regulation of CTB by mitochondrial fission regulator 1 like (MTFR1L) and the mechanisms. By studying human placenta samples and HTR-8/SVneo cell line, MTFR1L was found expressed in CTB. While MTFR1L expression was lower in PE placenta and CTB comparing with Normal pregnancy. Knockdown of MTFR1L decreased CTB invasion and migration, as well as the ATP production, while increased the mitochondria fragmentation, ROS production and mitochondria membrane potential indicating MTFR1L was key regulator of mitochondria. The posttranslational modulation analysis showed enhanced persulfidation of MTFR1L on cystine 222 and 230 by H2S. Mutations of MTFR1LC222/C230 suppressed ATP production, CTB invasion, migration, and increased mitochondria fragmentation, ROS production and mitochondria membrane potential. The present study showed the functional MTFR1L received endogenous CBS/H2S regulation. MTFR1LC222/230 persulfidation by H2S maintained mitochondria morphology and functions thus restored CTB invasion and migration. These findings established a new regulatory pathway for CTB invasion and migration, and provided new targets for PE treatment.
Collapse
Affiliation(s)
- Hao Feng
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Chunlei Tian
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Wenshan Jiang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250012, China
| | - Zongxin Sun
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Yikun Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Baoshi Han
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, Shandong 250012, China
| | - Lumei Chen
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Dawei Wang
- Central Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Hongjie Xiang
- Department of Traditional Chinese Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Jianchun Zhu
- Department of Pathology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Wengang Song
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Jingxin Li
- Department of Physiology, School of Basic Medical Sciences, Cheeloo College of Medicine, Shandong University, Jinan, Shandong 250012, China
| | - Yunlu Cai
- Laboratory of Metabolism and Gastrointestinal Tumor, The First Affiliated Hospital of Shandong First Medical University& Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China
| | - Shuanglian Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250012, China.
| | - Yan Li
- Shandong Province University Clinical Immunology Translational Medicine Laboratory, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China; Translational Medical Research Centre, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong 250014, China.
| |
Collapse
|
6
|
Carrillo R, Moreno I, Romero LC, Aroca A, Gotor C. Hydrogen sulfide-induced barley resilience to drought and salinity through protein persulfidation. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2025; 221:109644. [PMID: 39965413 DOI: 10.1016/j.plaphy.2025.109644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 01/15/2025] [Accepted: 02/10/2025] [Indexed: 02/20/2025]
Abstract
Barley (Hordeum vulgare) is a widely cultivated cereal crops, and its production is increasingly threatened by environmental stresses such as drought and salinity. Hydrogen sulfide is established as a signaling molecule that promotes tolerance to plant stress throught persulfidation, a post-translational modification of cysteine residues in proteins. The purpose of this study is to explore the impact of NaHS (sulfide donor) pretreatment on barley plants in enhancing tolerance to drought and salinity stresses, and determine if persulfidation is involved. In pretreated-plants, phenotypical traits and pigment contents showed an improvement in the survival of the plants under stress conditions. Quantification of stress-markers such as anthocyanin, proline, and reactive oxygen species also showed significant decreased contents in pretreated compared to untreated plants. In addition, the accumulation of amino acids under drought stress was significantly reduced when plants were pretreated with NaHS. Similarly, the increase of ABA content as a typical drought response was reduced in the pretreated plants. When plants are exposed to salt stress, the Na+/K+ ratio was maintained low in NaHS-pretreated plants, by increasing K+ levels. The sulfide ameliorative effect to salt was also observed during germination in previously NaHS-soaked seeds. Our findings suggest that sulfide pretreatment prepares barely plants to better deal with drought and salinity. Moreover, persulfidation was analyzed under all conditions, exhibiting enhanced levels under stress when plants were pretreated with NaHS. Our findings indicate that sulfide pretreatment induces a previous state in barley to respond more efficiently to stress and propose persulfidation is the underlying mechanism.
Collapse
Affiliation(s)
- Reyes Carrillo
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas and Universidad de Sevilla, Sevilla, Spain
| | - Inmaculada Moreno
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas and Universidad de Sevilla, Sevilla, Spain
| | - Luis C Romero
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas and Universidad de Sevilla, Sevilla, Spain
| | - Angeles Aroca
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas and Universidad de Sevilla, Sevilla, Spain.
| | - Cecilia Gotor
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas and Universidad de Sevilla, Sevilla, Spain.
| |
Collapse
|
7
|
Yang H, Ji X, Zhong H, Yang X, Hu D, Cai G, Wu Z. CRISPR screening identifies protein methylation and ubiquitination modifications that modulate aflatoxin B 1 cytotoxicity. SCIENCE CHINA. LIFE SCIENCES 2025:10.1007/s11427-024-2866-1. [PMID: 40172759 DOI: 10.1007/s11427-024-2866-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/11/2025] [Indexed: 04/04/2025]
Abstract
Aflatoxin B1 (AFB1) is one of the most potent mycotoxins affecting human health and animal production. To deeply understand the host-toxin interaction, we performed CRISPR screening and identified cystathionine β-synthase (CBS) as a critical host gene affecting AFB1 cytotoxicity. Mechanistic studies revealed that CBS affects AFB1-induced cell death by regulating the abundance of protein post-translational modifications (PTMs) in host cells. First, AFB1 disrupted the transfer of S-adenosylmethionine (SAM) from the cytoplasm to the mitochondria, thereby reducing the intra-mitochondrial protein methylation level. Deficient intra-mitochondrial protein methylation impaired mitochondrial function and caused cell death. CBS knockout (KO) can enhance SAM generation and mobilization to restore intra-mitochondrial SAM levels by rescuing the perturbed methionine cycle after AFB1 exposure, thereby alleviating AFB1-induced cell death. Second, AFB1 decreased global protein ubiquitination levels by affecting gene expression of ubiquitin-modified enzymes. CBS-KO and pharmaceutical treatment correcting gene expression of ubiquitin-modified enzymes can rescue AFB1-induced cell death. We also investigated two PTM-regulating small molecules, SAM and PR-619, which can increase cell viability in AFB1-exposed cells.
Collapse
Affiliation(s)
- Huaqiang Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu, 527400, China.
| | - Xi Ji
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Haiwen Zhong
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu, 527400, China
| | - Xiaohui Yang
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Dandan Hu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
| | - Gengyuan Cai
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu, 527400, China
| | - Zhenfang Wu
- State Key Laboratory of Swine and Poultry Breeding Industry, National Engineering Research Center for Breeding Swine Industry, College of Animal Science, South China Agricultural University, Guangzhou, 510642, China.
- Yunfu Branch Center of Guangdong Laboratory of Lingnan Modern Agricultural Science and Technology, Yunfu, 527400, China.
| |
Collapse
|
8
|
Khodade VS, Liu Q, Zhang C, Keceli G, Paolocci N, Toscano JP. Arylsulfonothioates: Thiol-Activated Donors of Hydropersulfides which are Excreted to Maintain Cellular Redox Homeostasis or Retained to Counter Oxidative Stress. J Am Chem Soc 2025; 147:7765-7776. [PMID: 39963866 DOI: 10.1021/jacs.4c17661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2025]
Abstract
Despite their biological significance, the study of hydropersulfides (RSSH) is often limited due to their inherent instability. Here, we introduce arylsulfonothioates as thiol-activated RSSH donors and provide insight into cellular reactive sulfur species homeostasis. These precursors persulfidate physiologically relevant thiols (RSH) to form the corresponding RSSH. Real-time monitoring of hydrogen sulfide (H2S) generation via membrane inlet mass spectrometry (MIMS) was employed to follow RSSH production, revealing that electron-donating aryl substituents marginally slow RSSH release rates, whereas electron-withdrawing substituents slightly accelerate release. Furthermore, arylsulfonothioates with strong electron-withdrawing substituents offer superior protection against doxorubicin (DOX)-induced cardiotoxicity. Experiments using H9c2 cardiomyocytes affirmed the cell-permeability of arylsulfonothioates and their ability to increase intracellular RSSH levels and protein persulfidation levels. Notably, we observe the excretion of RSSH into the extracellular medium. Further investigations revealed the involvement of the cystine/glutamate antiporter SLC7A11, as cotreatment with its inhibitor, sulfasalazine, significantly reduce extracellular RSSH release. H9c2 cells exhibit tolerance to arylsulfonothioate 1g with an electron-withdrawing 4-cyano group at 1 mM; however, inhibition of the cystine antiporter results in a minor decrease in cell viability. Under oxidative stress conditions induced by DOX or hydrogen peroxide (H2O2), cotreatment with 1g diminishes the excretion of RSSH and confers cytoprotection against DOX or H2O2-mediated toxicity. Our findings show adaptive cellular responses to RSSH levels, demonstrating excretion under elevated conditions to maintain redox homeostasis and intracellular retention as a protective response during oxidative stress.
Collapse
Affiliation(s)
- Vinayak S Khodade
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Qi Liu
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Chengximeng Zhang
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| | - Gizem Keceli
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
| | - Nazareno Paolocci
- Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, United States
- Department of Biomedical Sciences, University of Padova, Padova 35131, Italy
| | - John P Toscano
- Department of Chemistry, Johns Hopkins University, Baltimore, Maryland 21218, United States
| |
Collapse
|
9
|
Borbényi-Galambos K, Erdélyi K, Ditrói T, Jurányi EP, Szántó N, Szatmári R, Czikora Á, Schmidt EE, Garai D, Cserepes M, Liszkay G, Tóth E, Tóvári J, Nagy P. Realigned transsulfuration drives BRAF-V600E-targeted therapy resistance in melanoma. Cell Metab 2025:S1550-4131(25)00021-X. [PMID: 40037361 DOI: 10.1016/j.cmet.2025.01.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 12/13/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025]
Abstract
BRAF V600E-inhibition effectively treats melanoma, but acquired resistance rapidly develops. Protein expression profiles, mitochondrial energetics, metabolomics and fluxomics data in cell line, xenograft, and patient-derived xenograft systems revealed that concerted reprogramming of metabolic pathways (including glutaminolysis, glycolysis, TCA cycle, electron transport chain [ETC], and transsulfuration), along with an immediate cytoprotective response to drug-induced oxidative stress, underpins drug-tolerant persister cancer cell survival. Realignment of cysteine (Cys) metabolism, in particular an immediate upregulation of cystathionine-γ-lyase (CSE), was vital in persister cells. The oxidative cellular environment, drug-induced elevated cystine uptake and oxidative Cys catabolism, increased intracellular cystine/Cys ratios, thereby favoring cystine as a CSE substrate. This produces persulfides and hydrogen sulfide to protect protein thiols and support elevated energy demand in persister cells. Combining BRAF V600E inhibitors with CSE inhibitors effectively diminished proliferative relapse in culture models and increased progression-free survival of xenografted mice. This, together with induced CSE expression in patient samples under BRAF-V600E-inhibition, reveals an approach to increase BRAF-V600E-targeted therapeutic efficacy.
Collapse
Affiliation(s)
- Klaudia Borbényi-Galambos
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hajdú-Bihar County, 4032, Hungary
| | - Katalin Erdélyi
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Tamás Ditrói
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Eszter Petra Jurányi
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Semmelweis University Doctoral School, Semmelweis University, Budapest, 1094, Hungary
| | - Noémi Szántó
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Réka Szatmári
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hajdú-Bihar County, 4032, Hungary; Chemistry Coordinating Institute, University of Debrecen, Debrecen, Hajdú-Bihar County, 4012, Hungary
| | - Ágnes Czikora
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Edward E Schmidt
- Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology, University of Veterinary Medicine, Budapest, 1078, Hungary; Department of Microbiology and Cell Biology, Montana State University, Bozeman, Montana, 59717, United States of America
| | - Dorottya Garai
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Kálmán Laki Doctoral School, University of Debrecen, Debrecen, Hajdú-Bihar County, 4032, Hungary
| | - Mihály Cserepes
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Gabriella Liszkay
- Department of Dermatology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Erika Tóth
- Department of Surgical and Molecular Pathology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - József Tóvári
- Department of Experimental Pharmacology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary
| | - Péter Nagy
- Department of Molecular Immunology and Toxicology and the National Tumor Biology Laboratory, National Institute of Oncology, Budapest, 1122, Hungary; Chemistry Coordinating Institute, University of Debrecen, Debrecen, Hajdú-Bihar County, 4012, Hungary; Department of Anatomy and Histology, HUN-REN-UVMB Laboratory of Redox Biology, University of Veterinary Medicine, Budapest, 1078, Hungary.
| |
Collapse
|
10
|
Peng YJ, Nanduri J, Wang N, Su X, Hildreth M, Prabhakar NR. Signal Transduction Pathway Mediating Carotid Body Dependent Sympathetic Activation and Hypertension by Chronic Intermittent Hypoxia. FUNCTION 2025; 6:zqaf003. [PMID: 39844350 DOI: 10.1093/function/zqaf003] [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: 10/29/2024] [Revised: 01/03/2025] [Accepted: 01/18/2025] [Indexed: 01/24/2025] Open
Abstract
Patients with obstructive sleep apnea (OSA) experience chronic intermittent hypoxia (CIH). OSA patients and CIH-treated rodents exhibit overactive sympathetic nervous system and hypertension, mediated through hyperactive carotid body (CB) chemoreflex. Activation of olfactory receptor 78 (Olfr78) by hydrogen sulfide (H2S) is implicated in CB activation and sympathetic nerve responses to CIH, but the downstream signaling pathways remain unknown. Given that odorant receptor signaling is coupled to adenylyl cyclase 3 (Adcy3), we hypothesized that Adcy3-dependent cyclic adenosine monophosphate (cAMP) contributes to CB and sympathetic responses to CIH. Our findings show that CIH increases cAMP levels in the CB, a response absent in Adcy3, Cth (encoding CSE), and Olfr78 null mice. CBs from Cth and Olfr78 mutant mice lacked a persulfidation response to CIH, indicating that Adcy3 activation requires Olfr78 activation by H2S in CIH. CIH also enhanced glomus cell Ca2+ influx, an effect absent in Cnga2 (encoding cyclic nucleotide-gated channel alpha2 subunit) and Adcy3 mutants, suggesting that CIH-induced cAMP mediates enhanced Ca2+ responses through cyclic nucleotide-gated channels. Furthermore, Adcy3 null mice did not exhibit either CB activation or sympathetic activation by CIH. These results demonstrate that Adcy3-dependent cAMP is a downstream signaling pathway to H2S/Olfr78, mediating CIH-induced CB activation, sympathetic activity and hypertension.
Collapse
Affiliation(s)
- Ying-Jie Peng
- Institute for Integrative Physiology, Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Jayasri Nanduri
- Institute for Integrative Physiology, Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Ning Wang
- Institute for Integrative Physiology, Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Xiaoyu Su
- Institute for Integrative Physiology, Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Matthew Hildreth
- Institute for Integrative Physiology, Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| | - Nanduri R Prabhakar
- Institute for Integrative Physiology, Department of Medicine, Pritzker School of Medicine, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
11
|
Petrovic D, Slade L, Paikopoulos Y, D'Andrea D, Savic N, Stancic A, Miljkovic JL, Vignane T, Drekolia MK, Mladenovic D, Sutulovic N, Refeyton A, Kolakovic M, Jovanovic VM, Zivanovic J, Miler M, Vellecco V, Brancaleone V, Bucci M, Casey AM, Yu C, Kasarla SS, Smith KW, Kalfe-Yildiz A, Stenzel M, Miranda-Vizuete A, Hergenröder R, Phapale P, Stanojlovic O, Ivanovic-Burmazovic I, Vlaski-Lafarge M, Bibli SI, Murphy MP, Otasevic V, Filipovic MR. Ergothioneine improves healthspan of aged animals by enhancing cGPDH activity through CSE-dependent persulfidation. Cell Metab 2025; 37:542-556.e14. [PMID: 39842434 DOI: 10.1016/j.cmet.2024.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 09/22/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025]
Abstract
Ergothioneine (ET), a dietary thione/thiol, is receiving growing attention for its possible benefits in healthy aging and metabolic resilience. Our study investigates ET's effects on healthspan in aged animals, revealing lifespan extension and enhanced mobility in Caenorhabditis elegans, accompanied by improved stress resistance and reduced age-associated biomarkers. In aged rats, ET administration enhances exercise endurance, muscle mass, and vascularization, concomitant with higher NAD+ levels in muscle. Mechanistically, ET acts as an alternative substrate for cystathionine gamma-lyase (CSE), stimulating H2S production, which increases protein persulfidation of more than 300 protein targets. Among these, protein-persulfidation-driven activation of cytosolic glycerol-3-phosphate dehydrogenase (cGPDH) primarily contributes to the ET-induced NAD+ increase. ET's effects are abolished in models lacking CSE or cGPDH, highlighting the essential role of H2S signaling and protein persulfidation. These findings elucidate ET's multifaceted actions and provide insights into its therapeutic potential for combating age-related muscle decline and metabolic perturbations.
Collapse
Affiliation(s)
- Dunja Petrovic
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | - Luke Slade
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | | | - Davide D'Andrea
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | - Nevena Savic
- Institute for Biological Research "Sinisa Stankovic", National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Ana Stancic
- Institute for Biological Research "Sinisa Stankovic", National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jan Lj Miljkovic
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge CB2 0XY, UK
| | - Thibaut Vignane
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | - Maria Kyriaki Drekolia
- Department of Vascular Dysfunction, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dusan Mladenovic
- Institute for Pathophysiology "Ljubodrag Buba Mihailovic", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Nikola Sutulovic
- Laboratory for Neurophysiology, Institute for Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Alice Refeyton
- Inserm U1211 Maladies Rares: Génétique et Métabolisme, Université de Bordeaux, Bordeaux, France
| | - Milica Kolakovic
- Department of Chemistry, Ludwig Maximilians University of Munich, Munich, Germany
| | - Vladimir M Jovanovic
- Bioinformatics Solution Center, Institute for Informatics, Freie Universität Berlin, Berlin, Germany
| | - Jasmina Zivanovic
- Institute for Biological Research "Sinisa Stankovic", National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Marko Miler
- Institute for Biological Research "Sinisa Stankovic", National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Valentina Vellecco
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | | | - Mariarosaria Bucci
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy
| | - Alva M Casey
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge CB2 0XY, UK
| | - ChakShun Yu
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge CB2 0XY, UK
| | | | | | | | - Martin Stenzel
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | - Antonio Miranda-Vizuete
- Redox Homeostasis Group, Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Seville, Spain
| | | | - Prasad Phapale
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany
| | - Olivera Stanojlovic
- Laboratory for Neurophysiology, Institute for Medical Physiology "Richard Burian", Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | | | - Marija Vlaski-Lafarge
- Inserm U1211 Maladies Rares: Génétique et Métabolisme, Université de Bordeaux, Bordeaux, France
| | - Sofia-Iris Bibli
- Department of Vascular Dysfunction, European Center for Angioscience, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge CB2 0XY, UK; Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Vesna Otasevic
- Institute for Biological Research "Sinisa Stankovic", National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Milos R Filipovic
- Leibniz Institute for Analytical Sciences, ISAS e.V., Dortmund, Germany; School of Molecular Biosciences, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
12
|
Nishimura A, Tanaka T, Shimoda K, Ida T, Sasaki S, Umezawa K, Imamura H, Urano Y, Ichinose F, Kaneko T, Akaike T, Nishida M. Non-thermal atmospheric pressure plasma-irradiated cysteine protects cardiac ischemia/reperfusion injury by preserving supersulfides. Redox Biol 2025; 79:103445. [PMID: 39637599 PMCID: PMC11663985 DOI: 10.1016/j.redox.2024.103445] [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: 10/26/2024] [Revised: 11/26/2024] [Accepted: 11/26/2024] [Indexed: 12/07/2024] Open
Abstract
Ischemic heart disease is the main global cause of death in the world. Abnormal sulfide catabolism, especially hydrogen sulfide accumulation, impedes mitochondrial respiration and worsens the prognosis after ischemic insults, but the substantial therapeutic strategy has not been established. Non-thermal atmospheric pressure plasma irradiation therapy is attracted attention as it exerts beneficial effects by producing various reactive molecular species. Growing evidence has suggested that supersulfides, formed by catenation of sulfur atoms, contribute to various biological processes involving electron transfer in cells. Here, we report that non-thermal plasma-irradiated cysteine (Cys∗) protects mouse hearts against ischemia/reperfusion (I/R) injury by preventing supersulfide catabolism. Cys∗ has a weak but long-lasting supersulfide activity, and the treatment of rat cardiomyocytes with Cys∗ prevents mitochondrial dysfunction after hypoxic stress. Cys∗ increases sulfide-quinone oxidoreductase (SQOR), and silencing SQOR abolishes Cys∗-induced supersulfide formation and cytoprotection. Local administration of mouse hearts with Cys∗ significantly reduces infarct size with preserving supersulfide levels after I/R. These results suggest that maintaining supersulfide formation through SQOR underlies cardioprotection by Cys∗ against I/R injury.
Collapse
Affiliation(s)
- Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, 444-8787, Japan; SOKENDAI, Department of Physiological Sciences, Okazaki, 444-8787, Japan
| | - Tomohiro Tanaka
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, 444-8787, Japan; Center for Novel Science Initiatives (CNSI), National Institutes of Natural Sciences, Tokyo, 105-0001, Japan
| | - Kakeru Shimoda
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, 444-8787, Japan; SOKENDAI, Department of Physiological Sciences, Okazaki, 444-8787, Japan; Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Tomoaki Ida
- Organization for Research Promotion, Osaka Metropolitan University, Sakai, 599-8531, Japan; Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan
| | - Shota Sasaki
- Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Keitaro Umezawa
- Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, 173-0015, Japan
| | - Hiromi Imamura
- Organization of Research Initiatives, Yamaguchi University, Yamaguchi, 753-8515, Japan
| | - Yasuteru Urano
- Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo 113-0033, Japan; Graduate School of Medicine, The University of Tokyo, Tokyo, 113-0033, Japan
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Toshiro Kaneko
- Graduate School of Engineering, Tohoku University, Sendai, 980-8579, Japan
| | - Takaaki Akaike
- Graduate School of Medicine, Tohoku University, Sendai, 980-8575, Japan
| | - Motohiro Nishida
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Okazaki, 444-8787, Japan; SOKENDAI, Department of Physiological Sciences, Okazaki, 444-8787, Japan; Center for Novel Science Initiatives (CNSI), National Institutes of Natural Sciences, Tokyo, 105-0001, Japan; Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
13
|
Pan Y, Matsunaga T, Zhang T, Akaike T. The Therapeutic Potential of Supersulfides in Oxidative Stress-Related Diseases. Biomolecules 2025; 15:172. [PMID: 40001475 PMCID: PMC11852411 DOI: 10.3390/biom15020172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 01/10/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Oxidation-reduction (redox) reactions are fundamental to sustaining life, with reactive oxygen and nitrogen species playing pivotal roles in cellular signaling and homeostasis. However, excessive oxidative stress disrupts redox balance, contributing to a wide range of diseases, including inflammatory and pulmonary disorders, neurodegeneration, and cancer. Although numerous antioxidant therapies have been developed and tested for oxidative stress-related diseases, their clinical efficacy remains limited. Here, we introduce the emerging concept of 'supersulfides', a class of redox molecule species with unique antioxidant and nucleophilic properties, which have recently been recognized as crucial regulators of cellular redox homeostasis. Unlike traditional antioxidants, supersulfides offer novel mechanisms of action that directly target the underlying processes of oxidative stress. This review summarizes current knowledge on supersulfides, highlighting their roles in oxidative stress and associated diseases, as well as the mechanisms underlying oxidative stress-related pathology. The therapeutic potential of synthetic supersulfides for treating oxidative stress-related diseases is also discussed. A comprehensive understanding of the molecular and cellular basis of redox biology can help to guide the development of innovative redox-based therapeutic strategies aimed at preventing and treating diseases associated with disturbed redox regulation.
Collapse
Grants
- 20348438 Japan Science and Technology Agency
- 21H05263 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 21H05258 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 23K20040 Ministry of Education, Culture, Sports, Science and Technology of Japan
- 24H00063 Ministry of Education, Culture, Sports, Science and Technology of Japan
- JP21zf0127001 Japan Agency for Medical Research and Development
Collapse
Affiliation(s)
- Yuexuan Pan
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan;
| | - Tetsuro Matsunaga
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita 010-8543, Japan;
| | - Tianli Zhang
- Center for Integrated Control, Epidemiology and Molecular Pathophysiology of Infectious Diseases, Akita University, Akita 010-8543, Japan;
| | - Takaaki Akaike
- Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan;
| |
Collapse
|
14
|
Liu Z, Rouhier N, Couturier J. Dual Roles of Reducing Systems in Protein Persulfidation and Depersulfidation. Antioxidants (Basel) 2025; 14:101. [PMID: 39857435 PMCID: PMC11763069 DOI: 10.3390/antiox14010101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The oxidative modification of specific cysteine residues to persulfides is thought to be the main way by which hydrogen sulfide (H2S) exerts its biological and signaling functions. Therefore, protein persulfidation represents an important thiol-switching mechanism as other reversible redox post-translational modifications. Considering their reductase activity but also their connections with proteins that generate H2S and its related molecules, the glutaredoxin (GRX) and thioredoxin (TRX)-reducing systems have potential dual roles in both protein persulfidation and depersulfidation. In this review, we will first focus on recent advances describing the physiological pathways leading to protein persulfidation before discussing the dual roles of the physiological TRX and glutathione/GRX-reducing systems in protein persulfidation/depersulfidation.
Collapse
Affiliation(s)
- Zhichao Liu
- Université de Lorraine, INRAE, IAM, F-54000 Nancy, France
| | - Nicolas Rouhier
- Université de Lorraine, INRAE, IAM, F-54000 Nancy, France
- Institut Universitaire de France, F-75000 Paris, France
| | - Jérémy Couturier
- Université de Lorraine, INRAE, IAM, F-54000 Nancy, France
- Institut Universitaire de France, F-75000 Paris, France
| |
Collapse
|
15
|
Southern WM, Johnson EE, Fasbender EK, Fallon KS, Cavazos CL, Lowe DA, Rodney GG, Ervasti JM. Impaired hydrogen sulfide biosynthesis underlies eccentric contraction-induced force loss in dystrophin-deficient skeletal muscle. J Clin Invest 2025; 135:e176942. [PMID: 39808494 PMCID: PMC11870723 DOI: 10.1172/jci176942] [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: 10/25/2023] [Accepted: 01/08/2025] [Indexed: 01/16/2025] Open
Abstract
Eccentric contraction-induced (ECC-induced) force loss is a hallmark of murine dystrophin-deficient (mdx) skeletal muscle that is used to assess efficacy of potential therapies for Duchenne muscular dystrophy. While virtually all key proteins involved in muscle contraction have been implicated in ECC force loss, a unifying mechanism that orchestrates force loss across such diverse molecular targets has not been identified. We showed that correcting defective hydrogen sulfide (H2S) signaling in mdx muscle prevented ECC force loss. We also showed that the cysteine proteome of skeletal muscle functioned as a redox buffer in WT and mdx muscle during ECCs, but that buffer capacity in mdx muscle was significantly compromised by elevated basal protein oxidation. Finally, chemo-proteomic data suggested that H2S protected several proteins central to muscle contraction against irreversible oxidation through persulfidation-based priming. Our results support a unifying, redox-based mechanism of ECC force loss in mdx muscle.
Collapse
Affiliation(s)
- W. Michael Southern
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Erynn E. Johnson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Elizabeth K. Fasbender
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Katherine S. Fallon
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Courtney L. Cavazos
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - Dawn A. Lowe
- Department of Family Medicine and Community Health, Division of Physical Therapy and Rehabilitation Science, University of Minnesota, Minneapolis, Minnesota, USA
| | - George G. Rodney
- Department of Integrative Physiology, Baylor College of Medicine, Houston, Texas, USA
| | - James M. Ervasti
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
16
|
Vo ATT, Khan U, Liopo AV, Mouli K, Olson KR, McHugh EA, Tour JM, Pooparayil Manoj M, Derry PJ, Kent TA. Harshly Oxidized Activated Charcoal Enhances Protein Persulfidation with Implications for Neurodegeneration as Exemplified by Friedreich's Ataxia. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:2007. [PMID: 39728543 PMCID: PMC11728766 DOI: 10.3390/nano14242007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 12/28/2024]
Abstract
Harsh acid oxidation of activated charcoal transforms an insoluble carbon-rich source into water-soluble, disc structures of graphene decorated with multiple oxygen-containing functionalities. We term these pleiotropic nano-enzymes as "pleozymes". A broad redox potential spans many crucial redox reactions including the oxidation of hydrogen sulfide (H2S) to polysulfides and thiosulfate, dismutation of the superoxide radical (O2-*), and oxidation of NADH to NAD+. The oxidation of H2S is predicted to enhance protein persulfidation-the attachment of sulfur to cysteine residues. Persulfidated proteins act as redox intermediates, and persulfidation protects proteins from irreversible oxidation and ubiquitination, providing an important means of signaling. Protein persulfidation is believed to decline in several neurological disorders and aging. Importantly, and consistent with the role of persulfidation in signaling, the master antioxidant transcription factor Nrf2 is regulated by Keap1's persulfidation. Here, we demonstrate that pleozymes increased overall protein persulfidation in cells from apparently healthy individuals and from individuals with the mitochondrial protein mutation responsible for Friedreich's ataxia. We further find that pleozymes specifically enhanced Keap1 persulfidation, with subsequent increased accumulation of Nrf2 and Nrf2's antioxidant targets.
Collapse
Affiliation(s)
- Anh T. T. Vo
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | - Uffaf Khan
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | - Anton V. Liopo
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
- Department of Chemistry, Rice University, Houston, TX 77005, USA
| | - Karthik Mouli
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | - Kenneth R. Olson
- Department of Physiology, Indiana University School of Medicine South Bend, South Bend, IN 46617, USA
- Department of Biological Sciences, University of Notre Dame, Notre Dame, IN 46556, USA
| | - Emily A. McHugh
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- Smalley-Curl Institute, Rice University, Houston, TX 77005, USA
| | - James M. Tour
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- Smalley-Curl Institute, Rice University, Houston, TX 77005, USA
- Rice Advanced Materials Institute, Rice University, Houston, TX 77005, USA
- The NanoCarbon Center, Rice University, Houston, TX 77005, USA
| | - Madhavan Pooparayil Manoj
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
| | - Paul J. Derry
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
- School of Engineering Medicine, Texas A&M University, Houston, TX 77030, USA
| | - Thomas A. Kent
- Center for Genomics and Precision Medicine, Institute of Bioscience and Technology, Texas A&M Health Science Center, Houston, TX 77030, USA
- Department of Chemistry, Rice University, Houston, TX 77005, USA
- Stanley H. Appel Department of Neurology, Houston Methodist Hospital and Research Institute, Houston, TX 77030, USA
| |
Collapse
|
17
|
Liu H, Negoita F, Brook M, Sakamoto K, Morton NM. Quantification of persulfidation on specific proteins: are we nearly there yet? Essays Biochem 2024; 68:467-478. [PMID: 39290133 PMCID: PMC11625863 DOI: 10.1042/ebc20230095] [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: 04/19/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024]
Abstract
Hydrogen sulfide (H2S) played a pivotal role in the early evolution of life on Earth before the predominance of atmospheric oxygen. The legacy of a persistent role for H2S in life's processes recently emerged through its discovery in modern biochemistry as an endogenous cellular signalling modulator involved in numerous biological processes. One major mechanism through which H2S signals is protein cysteine persulfidation, an oxidative post-translational modification. In recent years, chemoproteomic technologies have been developed to allow the global scanning of protein persulfidation targets in mammalian cells and tissues, providing a powerful tool to elucidate the broader impact of altered H2S in organismal physiological health and human disease states. While hundreds of proteins were confirmed to be persulfidated by global persulfidome methodologies, the targeting of specific proteins of interest and the investigation of further mechanistic studies are still underdeveloped due to a lack of stringent specificity of the methods and the inherent instability of persulfides. This review provides an overview of the processes of endogenous H2S production, oxidation, and signalling and highlights the application and limitations of current persulfidation labelling approaches for investigation of this important evolutionarily conserved biological switch for protein function.
Collapse
Affiliation(s)
- Hongling Liu
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, U.K
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Florentina Negoita
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Matthew Brook
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, U.K
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen 2200, Denmark
| | - Nicholas M Morton
- Molecular Metabolism Group, University/BHF Centre for Cardiovascular Sciences, Queens Medical Research Institute, University of Edinburgh, U.K
- Centre for Systems Health and Integrated Metabolic Research, Department of Biosciences, School of Science and Technology, Nottingham Trent University, NG11 8NS, U.K
| |
Collapse
|
18
|
Chen T, Bai D, Gong C, Cao Y, Yan X, Peng R. Hydrogen sulfide mitigates mitochondrial dysfunction and cellular senescence in diabetic patients: Potential therapeutic applications. Biochem Pharmacol 2024; 230:116556. [PMID: 39332692 DOI: 10.1016/j.bcp.2024.116556] [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: 08/02/2024] [Revised: 09/08/2024] [Accepted: 09/23/2024] [Indexed: 09/29/2024]
Abstract
Diabetes induces a pro-aging state characterized by an increased abundance of senescent cells in various tissues, heightened chronic inflammation, reduced substance and energy metabolism, and a significant increase in intracellular reactive oxygen species (ROS) levels. This condition leads to mitochondrial dysfunction, including elevated oxidative stress, the accumulation of mitochondrial DNA (mtDNA) damage, mitophagy defects, dysregulation of mitochondrial dynamics, and abnormal energy metabolism. These dysfunctions result in intracellular calcium ion (Ca2+) homeostasis disorders, telomere shortening, immune cell damage, and exacerbated inflammation, accelerating the aging of diabetic cells or tissues. Hydrogen sulfide (H2S), a novel gaseous signaling molecule, plays a crucial role in maintaining mitochondrial function and mitigating the aging process in diabetic cells. This article systematically explores the specific mechanisms by which H2S regulates diabetes-induced mitochondrial dysfunction to delay cellular senescence, offering a promising new strategy for improving diabetes and its complications.
Collapse
Affiliation(s)
- Ting Chen
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Dacheng Bai
- Guangdong Institute of Mitochondrial Biomedicine, Room 501, Coolpad Building, No.2 Mengxi Road, High-tech Industrial Park, Nanshan District, Shenzhen, Guangdong Province 518000, China
| | - Changyong Gong
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Yu Cao
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China
| | - Xiaoqing Yan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China.
| | - Renyi Peng
- Institute of Life Sciences & Biomedicine Collaborative Innovation Center of Zhejiang Province, College of Life and Environmental Science, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
19
|
Kieronska-Rudek A, Ascencao K, Chlopicki S, Szabo C. Increased hydrogen sulfide turnover serves a cytoprotective role during the development of replicative senescence. Biochem Pharmacol 2024; 230:116595. [PMID: 39454733 DOI: 10.1016/j.bcp.2024.116595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/15/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
The mammalian gasotransmitter hydrogen sulfide (H2S) is produced by enzymes such as cystathionine β-synthase (CBS), cystathionine γ-lyase (CSE), 3-mercaptopyruvate sulfurtransferase (3-MST). Prior studies suggest that H2S may have cytoprotective and anti-aging effects. This project explores the regulation and role of endogenous H2S in a murine model of replicative senescence. H2S and polysulfide levels in RAW 264.7 murine macrophages (control cells: passage 5-10; senescent cells: passage 30-40) were measured using fluorescent probes. The expression of H2S-related enzymes and the activity of senescence marker beta-galactosidase (SA-β-Gal) were also analyzed. CBS, CSE, and 3-MST were inhibited using selective pharmacological inhibitors. Senescence led to a moderate upregulation of CBS and in a significant increase in CSE and 3-MST. H2S degradation enzymes were also elevated in senescence. Inhibition of H2S-producing enzymes reduced H2S levels but increased polysulfides. Inhibition of H2S production during senescence suppressed cell proliferation, and elevated SA-β-Gal and p21 levels. Comparing young and old mice spleens revealed downregulation of CBS and ETHE1 and upregulation of rhodanese and SUOX in older mice. The results demonstrate that increased reactive sulfur turnover occurs in senescent macrophages and that reactive sulfur species support cell proliferation and regulate cellular senescence.
Collapse
Affiliation(s)
- Anna Kieronska-Rudek
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Cracow, Poland
| | - Kelly Ascencao
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Stefan Chlopicki
- Jagiellonian University, Jagiellonian Centre for Experimental Therapeutics (JCET), Cracow, Poland; Jagiellonian University Medical College, Chair of Pharmacology, Faculty of Medicine, Cracow, Poland
| | - Csaba Szabo
- Chair of Pharmacology, Department of Science and Medicine, University of Fribourg, Fribourg, Switzerland.
| |
Collapse
|
20
|
Manna S, Agrawal R, Yadav T, Kumar TA, Kumari P, Dalai A, Kanade S, Balasubramanian N, Singh A, Chakrapani H. Orthogonal Persulfide Generation through Precision Tools Provides Insights into Mitochondrial Sulfane Sulfur. Angew Chem Int Ed Engl 2024; 63:e202411133. [PMID: 39091222 DOI: 10.1002/anie.202411133] [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: 06/28/2024] [Revised: 07/24/2024] [Accepted: 08/01/2024] [Indexed: 08/04/2024]
Abstract
The sulfane sulfur pool, comprised of persulfide (RS-SH) and polysulfide (RS-SnH) derived from hydrogen sulfide (H2S), has emerged as a major player in redox biochemistry. Mitochondria, besides energy generation, serve as significant cellular redox hubs, mediate stress response and cellular health. However, the effects of endogenous mitochondrial sulfane sulfur (MSS) remain largely uncharacterized as compared with their cytosolic counterparts, cytosolic sulfane sulfur (CSS). To investigate this, we designed a novel artificial substrate for mitochondrial 3-mercaptopyruvate sulfurtransferase (3-MST), a key enzyme involved in MSS biosynthesis. Using cells expressing a mitochondrion-localized persulfide biosensor, we demonstrate this tool's ability to selectively enhance MSS. While H2S was previously known to suppress human immunodeficiency virus (HIV-1), we found that MSS profoundly affected the HIV-1 life cycle, mediating viral reactivation from latency. Additionally, we provide evidence for the role of the host's mitochondrial redox state, membrane potential, apoptosis, and respiration rates in managing HIV-1 latency and reactivation. Together, dynamic fluctuations in the MSS pool have a significant and possibly conflicting effect on HIV-1 viral latency. The precision tools developed herein allow for orthogonal generation of persulfide within both mitochondria and the cytosol and will be useful in interrogating disease biology.
Collapse
Affiliation(s)
- Suman Manna
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Pune, Maharashtra, 411008, India
| | - Ragini Agrawal
- Department of Microbiology and Cell Biology, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Tarun Yadav
- Department of Biology, Indian Institute of Science Education and Research Pune, Pune, Maharashtra, 411008, India
| | - T Anand Kumar
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Pune, Maharashtra, 411008, India
| | - Pooja Kumari
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Pune, Maharashtra, 411008, India
| | - Aadishakti Dalai
- Department of Biology, Indian Institute of Science Education and Research Pune, Pune, Maharashtra, 411008, India
| | - Shaunak Kanade
- Department of Biology, Indian Institute of Science Education and Research Pune, Pune, Maharashtra, 411008, India
| | - Nagaraj Balasubramanian
- Department of Biology, Indian Institute of Science Education and Research Pune, Pune, Maharashtra, 411008, India
| | - Amit Singh
- Department of Microbiology and Cell Biology, Centre for Infectious Disease Research, Indian Institute of Science, Bangalore, Karnataka, 560012, India
| | - Harinath Chakrapani
- Department of Chemistry, Indian Institute of Science Education and Research Pune, Pune, Maharashtra, 411008, India
| |
Collapse
|
21
|
Hayashi M, Okazaki K, Papgiannakopoulos T, Motohashi H. The Complex Roles of Redox and Antioxidant Biology in Cancer. Cold Spring Harb Perspect Med 2024; 14:a041546. [PMID: 38772703 PMCID: PMC11529857 DOI: 10.1101/cshperspect.a041546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/23/2024]
Abstract
Redox reactions control fundamental biochemical processes, including energy production, metabolism, respiration, detoxification, and signal transduction. Cancer cells, due to their generally active metabolism for sustained proliferation, produce high levels of reactive oxygen species (ROS) compared to normal cells and are equipped with antioxidant defense systems to counteract the detrimental effects of ROS to maintain redox homeostasis. The KEAP1-NRF2 system plays a major role in sensing and regulating endogenous antioxidant defenses in both normal and cancer cells, creating a bivalent contribution of NRF2 to cancer prevention and therapy. Cancer cells hijack the NRF2-dependent antioxidant program and exploit a very unique metabolism as a trade-off for enhanced antioxidant capacity. This work provides an overview of redox metabolism in cancer cells, highlighting the role of the KEAP1-NRF2 system, selenoproteins, sulfur metabolism, heme/iron metabolism, and antioxidants. Finally, we describe therapeutic approaches that can be leveraged to target redox metabolism in cancer.
Collapse
Affiliation(s)
- Makiko Hayashi
- Department of Pathology, New York University School of Medicine, New York, New York 10016, USA
| | - Keito Okazaki
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| | | | - Hozumi Motohashi
- Department of Gene Expression Regulation, Institute of Development, Aging and Cancer, Tohoku University, Sendai 980-8575, Japan
| |
Collapse
|
22
|
Baeken MW. Sirtuins and their influence on autophagy. J Cell Biochem 2024; 125:e30377. [PMID: 36745668 DOI: 10.1002/jcb.30377] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2022] [Revised: 01/02/2023] [Accepted: 01/19/2023] [Indexed: 02/07/2023]
Abstract
Sirtuins and autophagy are well-characterized agents that can promote longevity and protect individual organisms from age-associated diseases like neurodegenerative disorders. In recent years, more and more data has been obtained that discerned potential overlaps and crosstalk between Sirtuin proteins and autophagic activity. This review aims to summarize the advances within the field for each individual Sirtuin in mammalian systems. In brief, most Sirtuins have been implicated in promoting autophagy, with Sirtuin 1 and Sirtuin 6 showing the highest immediate involvement, while Sirtuin 4 and Sirtuin 5 only demonstrate occasional influence. The way Sirtuins regulate autophagy, however, is very diverse, as they have been shown to regulate gene expression of autophagy-associated genes and posttranslational modifications of proteins, with consequences for the activity and cellular localization of these proteins. They have also been shown to determine specific proteins for autophagic degradation. Overall, much data has been accumulated over recent years, yet many open questions remain. Especially although the dynamic between Sirtuin proteins and the immediate regulation of autophagic players like Light Chain 3B has been confirmed, many of these proteins have various orthologues in mammalian systems, and research so far has not exceeded the bona fide components of autophagy.
Collapse
Affiliation(s)
- Marius W Baeken
- Nucleic Acid Chemistry and Engineering Unit, Okinawa Institute of Science and Technology Graduate University, Onna-son, Okinawa, Japan
| |
Collapse
|
23
|
He K, Tan B, Lu A, Bai L, Song C, Miao Y, Liu B, Chen Q, Teng X, Dai J, Wu Y. Asynchronous changes of hydrogen sulfide and its generating enzymes in most tissues with the aging process. Biosci Rep 2024; 44:BSR20240320. [PMID: 39312181 PMCID: PMC11473966 DOI: 10.1042/bsr20240320] [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: 03/11/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
Aging is an inevitable and irreversible biological process that gradually heightens the risks of various diseases and death. As a newly discovered endogenous gasotransmitter, hydrogen sulfide (H2S) has been identified to exert multiple beneficial impacts on the regulation of aging and age-related pathologies. This study was aimed at systematically exploring the relationship between asynchronous aging processes and H2S concentrations in various tissues of aging mice. Samples of plasma and 13 tissues were collected from four cross-sectional age groups (3, 6, 12 and 18 months of age) covering the lifespan of male C57BL/6J mice. The H2S concentration was quantified by a reported liquid chromatography-tandem mass spectrometry (LC-MS/MS) method with monobromobimane derivatization. Additionally, the expressions of cystathionine γ-lyase (CSE), cystathionine β-synthase and 3-mercaptopyruvate sulfurtransferase, in those tissues were analyzed by Western blotting. We discovered that the H2S concentrations decreased asynchronously with the aging process in plasma, heart, liver, kidney, spleen, subcutaneous fat and brown fat and increased in brain and lung. At least one of the three H2S-generating enzymes expressions was compensatorily up-regulated with the aging process in most tissues, among which the up-regulation of CSE was the most prominent.
Collapse
Affiliation(s)
- Kaichuan He
- Department of Physiology, Hebei Medical University, Hebei 050017, China
- Center for Clinical Medical Research, Hebei Genral Hospital, Hebei 050051, China
- Hebei Key Laboratory of Metabolic Diseases, Hebei Genral Hospital, Hebei 050051, China
| | - Bo Tan
- Clinical Pharmacokinetic Laboratory, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Ao Lu
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Lu Bai
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Chengqing Song
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Yuxin Miao
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Biyu Liu
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Qian Chen
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Xu Teng
- Department of Physiology, Hebei Medical University, Hebei 050017, China
| | - Jing Dai
- Department of Clinical Diagnostics, Hebei Medical University, Hebei 050017, China
| | - Yuming Wu
- Department of Physiology, Hebei Medical University, Hebei 050017, China
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, Hebei 050017, China
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang 050017, China
- Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, Shijiazhuang 050017, China
| |
Collapse
|
24
|
Feng H, Sun Z, Han B, Xia H, Chen L, Tian C, Yan S, Shi Y, Yin J, Song W, Gong P, Wang S, Li Y. Miro2 sulfhydration by CBS/H 2S promotes human trophoblast invasion and migration via regulating mitochondria dynamics. Cell Death Dis 2024; 15:776. [PMID: 39461943 PMCID: PMC11513031 DOI: 10.1038/s41419-024-07167-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 10/13/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024]
Abstract
Insufficient cytotrophoblast (CTB) migration and invasion into the maternal myometrium leads to pregnancy related complications like Intra-uterus Growth Restriction (IUGR), and pre-eclampsia (PE). We previously found that hydrogen sulfide (H2S) enhanced CTB migration without knowing the mechanism(s) and the pathophysiological significance. By studying human samples and cell line, we found that H2S levels were lower in PE patients' plasma; H2S synthetic enzyme cystathionine β-synthetase (CBS) was reduced in PE extravillious invasive trophoblasts. GYY4137 (H2S donor, 1 µM) promoted CBS/H2S translocation onto mitochondria, preserved mitochondria functions, enhanced cell invasion and migration. CBS knockdown hindered the above functions which were rescued by GYY4137, indicating the vital roles of CBS/H2S signal. Disturbance of mitochondria dynamics inhibited cell invasion and migration. The 185 and 504 cysteines of Mitochondrial Rho GTPase 2 (Miro2C185/C504) were highly sulfhydrated by H2S. Knockdown Miro2 or double mutation of Miro2C185/C504 to serine fragmented mitochondria, and inhibited cell invasion and migration which can't be rescued by H2S. The present study showed that human cytotrophoblast receives low dose H2S regulation; CBS/H2S sustained mitochondria functions via Miro2C185/C504 sulfhydration to enhance cytotrophoblast mobility. These findings established a new regulatory pathway for cytotrophoblast functions, and provided new targets for IUGR and PE.
Collapse
Affiliation(s)
- Hao Feng
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Zongxin Sun
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
- Department of Emergency, Affiliated Hospital of Chifeng University, Shifeng, 024001, China
| | - Baoshi Han
- Institute of Women, Children and Reproductive Health, Shandong University, Jinan, 250012, China
| | - Huitang Xia
- Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, Jinan, Shandong, 250014, PR China
| | - Lumei Chen
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Chunlei Tian
- Department of Obstetrics & Gynecology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Suhua Yan
- Department of Cardiology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Yugen Shi
- Department of Cardiology, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China
| | - Jie Yin
- Jinan Clinical Research Center for Tissue Engineering Skin Regeneration and Wound Repair, Jinan, Shandong, 250014, PR China
| | - Wengang Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250012, China
| | - Peipei Gong
- Department of Rehabilitation Medicine, the Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, China
| | - Shuanglian Wang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250012, China.
| | - Yan Li
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250012, China.
- Translational Medical Research Centre, the First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, 250014, China.
| |
Collapse
|
25
|
Zheng H, Chen H, Cai Y, Shen M, Li X, Han Y, Deng X, Cao H, Liu J, Li H, Liu B, Li G, Wang X, Chen H, Hou J, Lin SH, Zong L, Zhang Y. Hydrogen sulfide-mediated persulfidation regulates homocysteine metabolism and enhances ferroptosis in non-small cell lung cancer. Mol Cell 2024; 84:4016-4030.e6. [PMID: 39321805 DOI: 10.1016/j.molcel.2024.08.035] [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: 11/15/2023] [Revised: 06/15/2024] [Accepted: 08/30/2024] [Indexed: 09/27/2024]
Abstract
Hydrogen sulfide (H₂S), a metabolite of the transsulfuration pathway, has been implicated in ferroptosis, a unique form of cell death caused by lipid peroxidation. While the exact mechanisms controlling ferroptosis remain unclear, our study reveals that H₂S sensitizes human non-small cell lung cancer (NSCLC) cells to this process, particularly when cysteine levels are low. Combining H₂S with cystine depletion significantly enhances the effectiveness of ferroptosis-based cancer therapy. Mechanistically, H₂S persulfidates the 195th cysteine on S-adenosyl homocysteine hydrolase (SAHH), reducing its enzymatic activity. This leads to decreased homocysteine levels, subsequently lowering cysteine and glutathione concentrations under cystine depletion conditions. These changes ultimately increase the vulnerability of NSCLC cells to ferroptosis. Our findings establish H₂S as a key regulator of homocysteine metabolism and a critical factor in determining NSCLC cell susceptibility to ferroptosis. These results highlight the potential of H₂S-based therapies to improve the efficacy of ferroptosis-targeted cancer treatments for NSCLC.
Collapse
Affiliation(s)
- Hualei Zheng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Huidi Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yunjie Cai
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Min Shen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xilin Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yi Han
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xusheng Deng
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Hongjie Cao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Junjia Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Hao Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Benchao Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Ganlin Li
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Xindong Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Hui Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Jingjing Hou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, Fujian, China
| | - Shu-Hai Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute for Data Science in Health and Medicine Engineering, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, Fujian, China
| | - Lili Zong
- School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, Fujian, China
| | - Yongyou Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, Engineering Research Centre of Molecular Diagnostics of the Ministry of Education, School of Life Sciences, Xiamen University, Xiamen 361102, Fujian, China; National Institute for Data Science in Health and Medicine Engineering, Faculty of Medicine and Life Sciences, Shenzhen Research Institute of Xiamen University, Xiamen University, Xiamen 361102, Fujian, China.
| |
Collapse
|
26
|
Squarcina A, Maier P, Vignane T, Senft L, Filipovic MR, Ivanović-Burmazović I. Unlocking Selective Anticancer Mechanisms: Dinuclear Manganese Superoxide Dismutase Mimetics Combined with Pt(II) Complexes. Chemistry 2024; 30:e202402685. [PMID: 39037925 DOI: 10.1002/chem.202402685] [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: 07/15/2024] [Revised: 07/16/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
We conducted an in-depth exploration of the in vitro activities of the dinuclear Mn2L2Ac and Mn2L2 complexes (where HL=2-{[di(2-pyridyl)methylamino]-methyl}phenol), possessing dual superoxide dismutase (SOD) and catalase (CAT) activity. We investigated these complexes both individually and in conjunction with various Pt(II)-complexes, either as mixtures or as the Mn2-Pt adducts. Our findings revealed a notable up to 50 % enhancement in the viability of healthy human breast cells, contrasted with a viability decrease as low as 50 % in breast cancer cells upon combined treatments with Mn2 SOD mimics and Pt(II) complexes. Specifically, we synthesized and characterized the self-assembled Mn2-Pt adducts (isolated Mn2L2Pt and in situ Mn2L2Pt'), linking Mn2L2-core with the carboxylate group of PtDAPCl2 (dichloro(2,3-diaminopropionic acid) platinum(II)). The SOD activity of the isolated Mn2L2Pt adduct (kSOD=1.7×107 M-1 s-1) remained intact. Through in vitro cell viability assessments, ROS levels, cellular Mn uptake and proteomics measurements, we elucidated key mechanisms underlying the observed biological effects. We demonstrated that Mn2-containing formulations predominantly target mitochondrial processes, differently affecting the proteome of cancerous and healthy cells. They induced downregulation of H2S signaling and expression of mitochondrial complex I and III, as well as increased oxidative phosphorylation pathways and upregulation of EGFR in cancer cells. In contrast, healthy cells showed a decrease in EGFR expression and a moderate enrichment in oxidative phosphorylation pathways.
Collapse
Affiliation(s)
- Andrea Squarcina
- Department of Chemistry, Ludwig-Maximilians Universität (LMU) München, München, 81377, Germany
| | - Philipp Maier
- Department of Chemistry, Ludwig-Maximilians Universität (LMU) München, München, 81377, Germany
| | - Thibaut Vignane
- Leibniz Institute for Analytical Sciences ISAS e.V., Otto-Hahn-Straße 6b, 44227, Dortmund, Germany
| | - Laura Senft
- Department of Chemistry, Ludwig-Maximilians Universität (LMU) München, München, 81377, Germany
| | - Milos R Filipovic
- Leibniz Institute for Analytical Sciences ISAS e.V., Otto-Hahn-Straße 6b, 44227, Dortmund, Germany
| | | |
Collapse
|
27
|
Paul BD, Pieper AA. Neuroprotective signaling by hydrogen sulfide and its dysregulation in Alzheimer's disease. Curr Opin Chem Biol 2024; 82:102511. [PMID: 39142018 PMCID: PMC11390309 DOI: 10.1016/j.cbpa.2024.102511] [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: 06/02/2024] [Revised: 07/04/2024] [Accepted: 07/21/2024] [Indexed: 08/16/2024]
Abstract
The ancient messenger molecule hydrogen sulfide (H2S) modulates myriad signaling cascades and has been conserved across evolutionary boundaries. Although traditionally known as an environmental toxin, H2S is also synthesized endogenously to exert modulatory and homeostatic effects in a broad array of physiologic functions. Notably, H2S levels are tightly physiologically regulated, as both its excess and paucity can be toxic. Accumulating evidence has revealed pivotal roles for H2S in neuroprotection and normal cognitive function, and H2S homeostasis is dysregulated in neurodegenerative conditions. Here, we review the normal neuroprotective roles of H2S that go awry in Alzheimer's disease, the most common form of neurodegenerative disease.
Collapse
Affiliation(s)
- Bindu D Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Lieber Institute for Brain Development, Baltimore, MD, USA.
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA; Department of Neurosciences, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University, School of Medicine, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
28
|
Moseler A, Wagner S, Meyer AJ. Protein persulfidation in plants: mechanisms and functions beyond a simple stress response. Biol Chem 2024:hsz-2024-0038. [PMID: 39303198 DOI: 10.1515/hsz-2024-0038] [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: 03/01/2024] [Accepted: 08/28/2024] [Indexed: 09/22/2024]
Abstract
Posttranslational modifications (PTMs) can modulate the activity, localization and interactions of proteins and (re)define their biological function. Understanding how changing environments can alter cellular processes thus requires detailed knowledge about the dynamics of PTMs in time and space. A PTM that gained increasing attention in the last decades is protein persulfidation, where a cysteine thiol (-SH) is covalently bound to sulfane sulfur to form a persulfide (-SSH). The precise cellular mechanisms underlying the presumed persulfide signaling in plants are, however, only beginning to emerge. In the mitochondrial matrix, strict regulation of persulfidation and H2S homeostasis is of prime importance for maintaining mitochondrial bioenergetic processes because H2S is a highly potent poison for cytochrome c oxidase. This review summarizes the current knowledge about protein persulfidation and corresponding processes in mitochondria of the model plant Arabidopsis. These processes will be compared to the respective processes in non-plant models to underpin similarities or highlight apparent differences. We provide an overview of mitochondrial pathways that contribute to H2S and protein persulfide generation and mechanisms for H2S fixation and de-persulfidation. Based on current proteomic data, we compile a plant mitochondrial persulfidome and discuss how persulfidation may regulate protein function.
Collapse
Affiliation(s)
- Anna Moseler
- INRES-Chemical Signalling, University of Bonn, Friedrich-Ebert-Allee 144, D-53113 Bonn, Germany
| | - Stephan Wagner
- INRES-Chemical Signalling, University of Bonn, Friedrich-Ebert-Allee 144, D-53113 Bonn, Germany
| | - Andreas J Meyer
- INRES-Chemical Signalling, University of Bonn, Friedrich-Ebert-Allee 144, D-53113 Bonn, Germany
| |
Collapse
|
29
|
Pantaleno R, Scuffi D, Schiel P, Schwarzländer M, Costa A, García-Mata C. Mitochondrial ß-Cyanoalanine Synthase Participates in flg22-Induced Stomatal Immunity. PLANT, CELL & ENVIRONMENT 2024. [PMID: 39288437 DOI: 10.1111/pce.15155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 07/26/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
Plants regulate gas exchange with the environment and modulate transpirational water flow through guard cells, which set the aperture of the stomatal pores. External and internal stimuli are detected by guard cells and integrated into a signalling network that modulate turgor pressure and, hence, pore size. Pathogen-associated molecular patterns are among the stimuli that induce stomatal closure, to prevent pathogen entry through the pores, and this response, also referred to as stomatal immunity, is one of the hallmarks of PAMP-triggered immunity. While reactive oxygen species (ROS)-mediated signalling plays a key role in stomatal immunity, also the gasotransmitter hydrogen sulphide (H2S) interacts with key components of the guard cell signalling network to induce stomatal closure. While the role of H2S, produced by the main cytosolic source L-cysteine desulfhydrase 1, has been already investigated, there are additional enzymatic sources that synthesize H2S in different subcellular compartments. Their function has remained enigmatic, however. In this work, we elucidate the involvement of the mitochondrial H2S source, β-cyanoalanine synthase CAS-C1, on stomatal immunity induced by the bacterial PAMP flagellin (flg22). We show that cas-c1 plants are impaired to induce flg22-triggered stomatal closure and apoplastic ROS production, while they are more susceptible to bacterial surface inoculation. Moreover, mitochondrial H2S donor AP39 induced stomatal closure in an RBOHD-dependent manner, while depletion of endogenous H2S, impaired RBOHD-mediated apoplastic ROS production. In addition, pharmacological disruption of mitochondrial electron transport chain activity, affected stomatal closure produced by flg22, indicating its participation in the stomatal immunity response. Our findings add evidence to the emerging realization that intracellular organelles play a decisive role in orchestrating stomatal signalling and immune responses and suggest that mitochondrial-derived H2S is an important player of the stomatal immunity signalling network.
Collapse
Affiliation(s)
- Rosario Pantaleno
- Instituto de Investigaciones Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Denise Scuffi
- Instituto de Investigaciones Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Paula Schiel
- Instituto de Investigaciones Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| | - Markus Schwarzländer
- Institute of Plant Biology and Biotechnology (IBBP), University of Münster, Münster, Germany
| | - Alex Costa
- Department of Biosciences, University of Milan, Milan, Italy
| | - Carlos García-Mata
- Instituto de Investigaciones Biológicas, Consejo Nacional de Investigaciones Científicas y Técnicas, Universidad Nacional de Mar del Plata, Mar del Plata, Argentina
| |
Collapse
|
30
|
Liu RX, Song DK, Zhang YY, Gong HX, Jin YC, Wang XS, Jiang YL, Yan YX, Lu BN, Wu YM, Wang M, Li XB, Zhang K, Liu SB. L-Cysteine: A promising nutritional supplement for alleviating anxiety disorders. Neuroscience 2024; 555:213-221. [PMID: 39089569 DOI: 10.1016/j.neuroscience.2024.07.038] [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: 04/30/2024] [Revised: 07/19/2024] [Accepted: 07/23/2024] [Indexed: 08/04/2024]
Abstract
Anxiety disorders are prevalent chronic psychological disease with complex pathogenic mechanisms. Current anxiolytics have limited efficacy and numerous side effects in many anxiety patients, highlighting the urgent need for new therapies. Recent research has been focusing on nutritional supplements, particularly amino acids, as potential therapies for anxiety disorders. Among these, L-Cysteine plays a crucial role in various biological processes. L-Cysteine exhibits antioxidant properties that can enhance the antioxidant functions of the central nervous system (CNS). Furthermore, metabolites of L-cysteine, such as glutathione and hydrogen sulfide have been shown to alleviate anxiety through distinct molecular mechanisms. Long-term administration of L-Cysteine has anxiolytic, antidepressant, and memory-improving effects. L-Cysteine depletion can lead to increased oxidative stress in the brain. This review delves into the potential mechanisms of L-Cysteine and its main products, glutathione (GSH) and hydrogen sulfide (H2S) in the management of anxiety and related diseases.
Collapse
Affiliation(s)
- Rui-Xia Liu
- College of Life Sciences, Northwest University, Xi'an 710069, China; Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Da-Ke Song
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Ying-Ying Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Heng-Xin Gong
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yu-Chen Jin
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xin-Shang Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China
| | - Yong-Li Jiang
- Precision Pharmacy & Drug Development Center, Department of Pharmacy, Tangdu Hospital, Fourth Military Medical University, Xinsi Road 1, Shaanxi, Xi'an 710038, China
| | - Yu-Xuan Yan
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Bei-Ning Lu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Yu-Mei Wu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Min Wang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Xu-Bo Li
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China
| | - Kun Zhang
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China.
| | - Shui-Bing Liu
- Department of Pharmacology, School of Pharmacy, Fourth Military Medical University, Xi'an 710032, China; State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Pharmacy, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
31
|
Pilsova Z, Pilsova A, Zelenkova N, Klusackova B, Chmelikova E, Postlerova P, Sedmikova M. Hydrogen sulfide and its potential as a possible therapeutic agent in male reproduction. Front Endocrinol (Lausanne) 2024; 15:1427069. [PMID: 39324123 PMCID: PMC11423738 DOI: 10.3389/fendo.2024.1427069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/21/2024] [Indexed: 09/27/2024] Open
Abstract
Hydrogen sulfide (H2S) is an endogenously produced signaling molecule that belongs to the group of gasotransmitters along with nitric oxide (NO) and carbon monoxide (CO). H2S plays a pivotal role in male reproductive processes. It is produced in various tissues and cells of the male reproductive system, including testicular tissue, Leydig and Sertoli cells, epididymis, seminal plasma, prostate, penile tissues, and sperm cells. This review aims to summarize the knowledge about the presence and effects of H2S in male reproductive tissues and outline possible therapeutic strategies in pathological conditions related to male fertility, e. g. spermatogenetic disorders and erectile dysfunction (ED). For instance, H2S supports spermatogenesis by maintaining the integrity of the blood-testicular barrier (BTB), stimulating testosterone production, and providing cytoprotective effects. In spermatozoa, H2S modulates sperm motility, promotes sperm maturation, capacitation, and acrosome reaction, and has significant cytoprotective effects. Given its vasorelaxant effects, it supports the erection of penile tissue. These findings suggest the importance and therapeutic potential of H2S in male reproduction, paving the way for further research and potential clinical applications.
Collapse
Affiliation(s)
- Zuzana Pilsova
- Department of Veterinary Sciences, Faculty of Agrobiology, Food, and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| | - Aneta Pilsova
- Department of Veterinary Sciences, Faculty of Agrobiology, Food, and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| | - Natalie Zelenkova
- Department of Veterinary Sciences, Faculty of Agrobiology, Food, and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| | - Barbora Klusackova
- Department of Veterinary Sciences, Faculty of Agrobiology, Food, and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| | - Eva Chmelikova
- Department of Veterinary Sciences, Faculty of Agrobiology, Food, and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| | - Pavla Postlerova
- Department of Veterinary Sciences, Faculty of Agrobiology, Food, and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
- Laboratory of Reproductive Biology, Institute of Biotechnology of the Czech Academy of Sciences, BIOCEV, Vestec, Czechia
| | - Marketa Sedmikova
- Department of Veterinary Sciences, Faculty of Agrobiology, Food, and Natural Resources, Czech University of Life Sciences Prague, Prague, Czechia
| |
Collapse
|
32
|
Qiu B, Boudker O. Structural basis of the excitatory amino acid transporter 3 substrate recognition. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.05.611541. [PMID: 39282329 PMCID: PMC11398500 DOI: 10.1101/2024.09.05.611541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/20/2024]
Abstract
Excitatory amino acid transporters (EAATs) reside on cell surfaces and uptake substrates, including L-glutamate, L-aspartate, and D-aspartate, using ion gradients. Among five EAATs, EAAT3 is the only isoform that can efficiently transport L-cysteine, a substrate for glutathione synthesis. Recent work suggests that EAAT3 also transports the oncometabolite R-2-hydroxyglutarate (R-2HG). Here, we examined the structural basis of substrate promiscuity by determining the cryo-EM structures of EAAT3 bound to different substrates. We found that L-cysteine binds to EAAT3 in thiolate form, and EAAT3 recognizes different substrates by fine-tuning local conformations of the coordinating residues. However, using purified human EAAT3, we could not observe R-2HG binding or transport. Imaging of EAAT3 bound to L-cysteine revealed several conformational states, including an outward-facing state with a semi-open gate and a disrupted sodium-binding site. These structures illustrate that the full gate closure, coupled with the binding of the last sodium ion, occurs after substrate binding. Furthermore, we observed that different substrates affect how the transporter distributes between a fully outward-facing conformation and intermediate occluded states on a path to the inward-facing conformation, suggesting that translocation rates are substrate-dependent.
Collapse
Affiliation(s)
- Biao Qiu
- Department of Physiology & Biophysics, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
- Howard Hughes Medical Institute, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
| | - Olga Boudker
- Department of Physiology & Biophysics, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
- Howard Hughes Medical Institute, Weill Cornell Medicine, 1300 York Ave, New York, NY 10021, USA
| |
Collapse
|
33
|
Jin Y, Yuan H, Liu Y, Zhu Y, Wang Y, Liang X, Gao W, Ren Z, Ji X, Wu D. Role of hydrogen sulfide in health and disease. MedComm (Beijing) 2024; 5:e661. [PMID: 39156767 PMCID: PMC11329756 DOI: 10.1002/mco2.661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 08/20/2024] Open
Abstract
In the past, hydrogen sulfide (H2S) was recognized as a toxic and dangerous gas; in recent years, with increased research, we have discovered that H2S can act as an endogenous regulatory transmitter. In mammals, H2S-catalyzing enzymes, such as cystathionine-β-synthase, cystathionine-γ-lyase, and 3-mercaptopyruvate sulfurtransferase, are differentially expressed in a variety of tissues and affect a variety of biological functions, such as transcriptional and posttranslational modification of genes, activation of signaling pathways in the cell, and metabolic processes in tissues, by producing H2S. Various preclinical studies have shown that H2S affects physiological and pathological processes in the body. However, a detailed systematic summary of these roles in health and disease is lacking. Therefore, this review provides a thorough overview of the physiological roles of H2S in different systems and the diseases associated with disorders of H2S metabolism, such as ischemia-reperfusion injury, hypertension, neurodegenerative diseases, inflammatory bowel disease, and cancer. Meanwhile, this paper also introduces H2S donors and novel release modes, as well as the latest preclinical experimental results, aiming to provide researchers with new ideas to discover new diagnostic targets and therapeutic options.
Collapse
Affiliation(s)
- Yu‐Qing Jin
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Hang Yuan
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Ya‐Fang Liu
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Yi‐Wen Zhu
- School of Clinical MedicineHenan UniversityKaifengHenanChina
| | - Yan Wang
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Xiao‐Yi Liang
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Wei Gao
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Zhi‐Guang Ren
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
| | - Xin‐Ying Ji
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
- Faculty of Basic Medical SubjectsShu‐Qing Medical College of ZhengzhouZhengzhouHenanChina
| | - Dong‐Dong Wu
- Henan International Joint Laboratory for Nuclear Protein RegulationSchool of Basic Medical Sciences, School of StomatologyHenan UniversityKaifengHenanChina
- School of StomatologyHenan UniversityKaifengHenanChina
- Department of StomatologyHuaihe Hospital of Henan UniversityKaifengHenanChina
| |
Collapse
|
34
|
Romanelli-Cedrez L, Vairoletti F, Salinas G. Rhodoquinone-dependent electron transport chain is essential for Caenorhabditis elegans survival in hydrogen sulfide environments. J Biol Chem 2024; 300:107708. [PMID: 39178951 PMCID: PMC11422605 DOI: 10.1016/j.jbc.2024.107708] [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: 02/28/2024] [Revised: 07/29/2024] [Accepted: 08/14/2024] [Indexed: 08/26/2024] Open
Abstract
Hydrogen sulfide (H2S) has traditionally been considered an environmental toxin for animal lineages; yet, it plays a signaling role in various processes at low concentrations. Mechanisms controlling H2S in animals, especially in sulfide-rich environments, are not fully understood. The main detoxification pathway involves the conversion of H2S into less harmful forms, through a mitochondrial oxidation pathway. The first step of this pathway oxidizes sulfide and reduces ubiquinone (UQ) through sulfide-quinone oxidoreductase (SQRD/SQOR). Because H2S inhibits cytochrome oxidase and hence UQ regeneration, this pathway becomes compromised at high H2S concentrations. The free-living nematode Caenorhabditis elegans feeds on bacteria and can face high sulfide concentrations in its natural environment. This organism has an alternative ETC that uses rhodoquinone (RQ) as the lipidic electron transporter and fumarate as the final electron acceptor. In this study, we demonstrate that RQ is essential for survival in sulfide. RQ-less animals (kynu-1 and coq-2e KO) cannot survive high H2S concentrations, while UQ-less animals (clk-1 and coq-2a KO) exhibit recovery, even when provided with a UQ-deficient diet. Our findings highlight that sqrd-1 uses both benzoquinones and that RQ-dependent ETC confers a key advantage (RQ regeneration) over UQ in sulfide-rich conditions. C. elegans also faces cyanide, another cytochrome oxidase inhibitor, whose detoxification leads to H2S production, via cysl-2. Our study reveals that RQ delays killing by the HCN-producing bacteria Pseudomonas aeruginosa PAO1. These results underscore the fundamental role that RQ-dependent ETC serves as a biochemical adaptation to H2S environments, and to pathogenic bacteria producing cyanide and H2S toxins.
Collapse
Affiliation(s)
| | - Franco Vairoletti
- Worm Biology Lab, Institut Pasteur de Montevideo, Montevideo, Uruguay; Laboratorio de Química Farmacéutica, Departamento de Química Orgánica, Facultad de Química, Universidad de la República, Montevideo, Uruguay
| | - Gustavo Salinas
- Worm Biology Lab, Institut Pasteur de Montevideo, Montevideo, Uruguay; Departamento de Biociencias, Facultad de Química, Universidad de la República, Montevideo, Uruguay.
| |
Collapse
|
35
|
Berndt C, Alborzinia H, Amen VS, Ayton S, Barayeu U, Bartelt A, Bayir H, Bebber CM, Birsoy K, Böttcher JP, Brabletz S, Brabletz T, Brown AR, Brüne B, Bulli G, Bruneau A, Chen Q, DeNicola GM, Dick TP, Distéfano A, Dixon SJ, Engler JB, Esser-von Bieren J, Fedorova M, Friedmann Angeli JP, Friese MA, Fuhrmann DC, García-Sáez AJ, Garbowicz K, Götz M, Gu W, Hammerich L, Hassannia B, Jiang X, Jeridi A, Kang YP, Kagan VE, Konrad DB, Kotschi S, Lei P, Le Tertre M, Lev S, Liang D, Linkermann A, Lohr C, Lorenz S, Luedde T, Methner A, Michalke B, Milton AV, Min J, Mishima E, Müller S, Motohashi H, Muckenthaler MU, Murakami S, Olzmann JA, Pagnussat G, Pan Z, Papagiannakopoulos T, Pedrera Puentes L, Pratt DA, Proneth B, Ramsauer L, Rodriguez R, Saito Y, Schmidt F, Schmitt C, Schulze A, Schwab A, Schwantes A, Soula M, Spitzlberger B, Stockwell BR, Thewes L, Thorn-Seshold O, Toyokuni S, Tonnus W, Trumpp A, Vandenabeele P, Vanden Berghe T, Venkataramani V, Vogel FCE, von Karstedt S, Wang F, Westermann F, Wientjens C, Wilhelm C, Wölk M, Wu K, Yang X, Yu F, Zou Y, Conrad M. Ferroptosis in health and disease. Redox Biol 2024; 75:103211. [PMID: 38908072 PMCID: PMC11253697 DOI: 10.1016/j.redox.2024.103211] [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: 03/21/2024] [Revised: 05/24/2024] [Accepted: 05/24/2024] [Indexed: 06/24/2024] Open
Abstract
Ferroptosis is a pervasive non-apoptotic form of cell death highly relevant in various degenerative diseases and malignancies. The hallmark of ferroptosis is uncontrolled and overwhelming peroxidation of polyunsaturated fatty acids contained in membrane phospholipids, which eventually leads to rupture of the plasma membrane. Ferroptosis is unique in that it is essentially a spontaneous, uncatalyzed chemical process based on perturbed iron and redox homeostasis contributing to the cell death process, but that it is nonetheless modulated by many metabolic nodes that impinge on the cells' susceptibility to ferroptosis. Among the various nodes affecting ferroptosis sensitivity, several have emerged as promising candidates for pharmacological intervention, rendering ferroptosis-related proteins attractive targets for the treatment of numerous currently incurable diseases. Herein, the current members of a Germany-wide research consortium focusing on ferroptosis research, as well as key external experts in ferroptosis who have made seminal contributions to this rapidly growing and exciting field of research, have gathered to provide a comprehensive, state-of-the-art review on ferroptosis. Specific topics include: basic mechanisms, in vivo relevance, specialized methodologies, chemical and pharmacological tools, and the potential contribution of ferroptosis to disease etiopathology and progression. We hope that this article will not only provide established scientists and newcomers to the field with an overview of the multiple facets of ferroptosis, but also encourage additional efforts to characterize further molecular pathways modulating ferroptosis, with the ultimate goal to develop novel pharmacotherapies to tackle the various diseases associated with - or caused by - ferroptosis.
Collapse
Affiliation(s)
- Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Hamed Alborzinia
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Vera Skafar Amen
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Scott Ayton
- Florey Institute of Neuroscience and Mental Health, University of Melbourne, Australia
| | - Uladzimir Barayeu
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany; Department of Environmental Medicine and Molecular Toxicology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Alexander Bartelt
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany; Institute for Diabetes and Cancer (IDC), Helmholtz Center Munich, Neuherberg, Germany; German Center for Cardiovascular Research, Partner Site Munich Heart Alliance, Munich, Germany
| | - Hülya Bayir
- Department of Pediatrics, Columbia University, New York City, NY, USA
| | - Christina M Bebber
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany
| | - Kivanc Birsoy
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Jan P Böttcher
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | - Simone Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Thomas Brabletz
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Ashley R Brown
- Department of Biological Sciences, Columbia University, New York City, NY, USA
| | - Bernhard Brüne
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Giorgia Bulli
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany
| | - Alix Bruneau
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Quan Chen
- College of Life Sciences, Nankai University, Tianjin, China
| | - Gina M DeNicola
- Department of Metabolism and Physiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Tobias P Dick
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ) Heidelberg, Germany; Faculty of Biosciences, Heidelberg University, 69120, Heidelberg, Germany
| | - Ayelén Distéfano
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Scott J Dixon
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Jan B Engler
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | | | - Maria Fedorova
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - José Pedro Friedmann Angeli
- Rudolf Virchow Zentrum, Center for Integrative and Translational Bioimaging - University of Würzburg, Germany
| | - Manuel A Friese
- Institute of Neuroimmunology and Multiple Sclerosis, University Medical Center Hamburg-Eppendorf, Germany
| | - Dominic C Fuhrmann
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Ana J García-Sáez
- Institute for Genetics, CECAD, University of Cologne, Germany; Max Planck Institute of Biophysics, Frankfurt/Main, Germany
| | | | - Magdalena Götz
- Department of Physiological Genomics, Ludwig-Maximilians-University, Munich, Germany; Institute of Stem Cell Research, Helmholtz Center Munich, Germany
| | - Wei Gu
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Linda Hammerich
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow-Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | | | - Xuejun Jiang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Aicha Jeridi
- Institute of Lung Health and Immunity (LHI), Helmholtz Munich, Comprehensive Pneumology Center (CPC-M), Germany, Member of the German Center for Lung Research (DZL)
| | - Yun Pyo Kang
- College of Pharmacy and Research Institute of Pharmaceutical Science, Seoul National University, Republic of Korea
| | | | - David B Konrad
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Stefan Kotschi
- Institute for Cardiovascular Prevention (IPEK), Faculty of Medicine, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Peng Lei
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Marlène Le Tertre
- Center for Translational Biomedical Iron Research, Heidelberg University, Germany
| | - Sima Lev
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Deguang Liang
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York City, NY, USA
| | - Andreas Linkermann
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany; Division of Nephrology, Department of Medicine, Albert Einstein College of Medicine, New York, NY, USA
| | - Carolin Lohr
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Svenja Lorenz
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Tom Luedde
- Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Axel Methner
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Bernhard Michalke
- Research Unit Analytical Biogeochemistry, Helmholtz Center Munich, Germany
| | - Anna V Milton
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Junxia Min
- School of Medicine, Zhejiang University, Hangzhou, China
| | - Eikan Mishima
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | | | - Hozumi Motohashi
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | | | - Shohei Murakami
- Department of Gene Expression Regulation, Tohoku University, Sendai, Japan
| | - James A Olzmann
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA; Department of Nutritional Sciences and Toxicology, University of California, Berkeley, CA, USA; Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Gabriela Pagnussat
- Instituto de Investigaciones Biológicas, CONICET, National University of Mar Del Plata, Argentina
| | - Zijan Pan
- School of Life Sciences, Westlake University, Hangzhou, China
| | | | | | - Derek A Pratt
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Canada
| | - Bettina Proneth
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany
| | - Lukas Ramsauer
- Institute of Molecular Immunology, School of Medicine, Technical University of Munich (TUM), Germany
| | | | - Yoshiro Saito
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Felix Schmidt
- Institute of Molecular Medicine, Johannes Gutenberg-Universität Mainz, Germany
| | - Carina Schmitt
- Department of Pharmacy, Ludwig-Maximilians-University, Munich, Germany
| | - Almut Schulze
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Annemarie Schwab
- Department of Experimental Medicine 1, Nikolaus-Fiebiger Center for Molecular Medicine, Friedrich-Alexander University of Erlangen-Nürnberg, Germany
| | - Anna Schwantes
- Institute of Biochemistry1-Pathobiochemistry, Goethe-Universität, Frankfurt Am Main, Germany
| | - Mariluz Soula
- Laboratory of Metabolic Regulation and Genetics, Rockefeller University, New York City, NY, USA
| | - Benedikt Spitzlberger
- Department of Immunobiology, Université de Lausanne, Switzerland; Center of Allergy and Environment (ZAUM), Technical University of Munich and Helmholtz Center Munich, Munich, Germany
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York City, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA; Department of Chemistry, Columbia University, New York, NY, USA
| | - Leonie Thewes
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | | | - Shinya Toyokuni
- Department of Pathology and Biological Responses, Nagoya University Graduate School of Medicine, Nagoya, Japan; Center for Low-temperature Plasma Sciences, Nagoya University, Nagoya, Japan; Center for Integrated Sciences of Low-temperature Plasma Core Research (iPlasma Core), Tokai National Higher Education and Research System, Nagoya, Japan
| | - Wulf Tonnus
- Division of Nephrology, Department of Internal Medicine III, University Hospital Carl Gustav Carus at the Technische Universität Dresden, Germany
| | - Andreas Trumpp
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM GGmbH), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany; German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Peter Vandenabeele
- VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Tom Vanden Berghe
- Department of Biomedical Sciences, University of Antwerp, Belgium; VIB-UGent Center for Inflammation Research, Ghent, Belgium; Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Vivek Venkataramani
- Comprehensive Cancer Center Mainfranken, University Hospital Würzburg, Germany
| | - Felix C E Vogel
- Division of Tumour Metabolism and Microenvironment, DKFZ Heidelberg and DKFZ-ZMBH Alliance, Heidelberg, Germany
| | - Silvia von Karstedt
- University of Cologne, Faculty of Medicine and University Hospital Cologne, Department of Translational Genomics, Cologne, Germany; CECAD Cluster of Excellence, University of Cologne, Cologne, Germany; University of Cologne, Faculty of Medicine and University Hospital Cologne, Center for Molecular Medicine Cologne, Germany
| | - Fudi Wang
- School of Medicine, Zhejiang University, Hangzhou, China
| | | | - Chantal Wientjens
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Christoph Wilhelm
- Immunopathology Unit, Institute of Clinical Chemistry and Clinical Pharmacology, Medical Faculty, University Hospital Bonn, University of Bonn, Germany
| | - Michele Wölk
- Center of Membrane Biochemistry and Lipid Research, University Hospital Carl Gustav Carus and Faculty of Medicine of TU Dresden, Germany
| | - Katherine Wu
- Department of Pathology, Grossman School of Medicine, New York University, NY, USA
| | - Xin Yang
- Institute for Cancer Genetics, And Herbert Irving Comprehensive Cancer Center, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Fan Yu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Yilong Zou
- School of Life Sciences, Westlake University, Hangzhou, China; Westlake Four-Dimensional Dynamic Metabolomics (Meta4D) Laboratory, Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Center Munich, Germany.
| |
Collapse
|
36
|
Slade L, Etheridge T, Szewczyk NJ. Consolidating multiple evolutionary theories of ageing suggests a need for new approaches to study genetic contributions to ageing decline. Ageing Res Rev 2024; 100:102456. [PMID: 39153601 DOI: 10.1016/j.arr.2024.102456] [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: 03/10/2024] [Revised: 08/05/2024] [Accepted: 08/13/2024] [Indexed: 08/19/2024]
Abstract
Understanding mechanisms of ageing remains a complex challenge for biogerontologists, but recent adaptations of evolutionary ageing theories offer a compelling lens in which to view both age-related molecular and physiological deterioration. Ageing is commonly associated with progressive declines in biochemical and molecular processes resulting from damage accumulation, yet the role of continued developmental gene activation is less appreciated. Natural selection pressures are at their highest in youthful periods to modify gene expression towards maximising reproductive capacity. After sexual maturation, selective pressure diminishes, subjecting individuals to maladaptive pleiotropic gene functions that were once beneficial for developmental growth but become pathogenic later in life. Due to this selective 'shadowing' in ageing, mechanisms to counter such hyper/hypofunctional genes are unlikely to evolve. Interventions aimed at targeting gene hyper/hypofunction during ageing might, therefore, represent an attractive therapeutic strategy. The nematode Caenorhabditis elegans offers a strong model for post-reproductive mechanistic and therapeutic investigations, yet studies examining the mechanisms of, and countermeasures against, ageing decline largely intervene from larval stages onwards. Importantly, however, lifespan extending conditions frequently impair early-life fitness and fail to correspondingly increase healthspan. Here, we consolidate multiple evolutionary theories of ageing and discuss data supporting hyper/hypofunctional changes at a global molecular and functional level in C. elegans, and how classical lifespan-extension mutations alter these dynamics. The relevance of such mutant models for exploring mechanisms of ageing are discussed, highlighting that post-reproductive gene optimisation represents a more translatable approach for C. elegans research that is not constrained by evolutionary trade-offs. Where some genetic mutations in C. elegans that promote late-life health map accordingly with healthy ageing in humans, other widely used genetic mutations that extend worm lifespan are associated with life-limiting pathologies in people. Lifespan has also become the gold standard for quantifying 'ageing', but we argue that gerospan compression (i.e., 'healthier' ageing) is an appropriate goal for anti-ageing research, the mechanisms of which appear distinct from those regulating lifespan alone. There is, therefore, an evident need to re-evaluate experimental approaches to study the role of hyper/hypofunctional genes in ageing in C. elegans.
Collapse
Affiliation(s)
- Luke Slade
- University of Exeter Medical School, Exeter, UK.
| | - Timothy Etheridge
- Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Nathaniel J Szewczyk
- Ohio Musculoskeletal and Neurological Institute, Heritage College of Osteopathic Medicine, Athens, OH 45701, United States.
| |
Collapse
|
37
|
Wang RH, Chen PR, Chen YT, Chen YC, Chu YH, Chien CC, Chien PC, Lo SY, Wang ZL, Tsou MC, Chen SY, Chiu GS, Chen WL, Wu YH, Wang LHC, Wang WC, Lin SY, Kung HJ, Wang LH, Cheng HC, Lin KT. Hydrogen sulfide coordinates glucose metabolism switch through destabilizing tetrameric pyruvate kinase M2. Nat Commun 2024; 15:7463. [PMID: 39198443 PMCID: PMC11358145 DOI: 10.1038/s41467-024-51875-9] [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: 09/02/2023] [Accepted: 08/20/2024] [Indexed: 09/01/2024] Open
Abstract
Most cancer cells reprogram their glucose metabolic pathway from oxidative phosphorylation to aerobic glycolysis for energy production. By reducing enzyme activity of pyruvate kinase M2 (PKM2), cancer cells attain a greater fraction of glycolytic metabolites for macromolecule synthesis needed for rapid proliferation. Here we demonstrate that hydrogen sulfide (H2S) destabilizes the PKM2 tetramer into monomer/dimer through sulfhydration at cysteines, notably at C326, leading to reduced PKM2 enzyme activity and increased PKM2-mediated transcriptional activation. Blocking PKM2 sulfhydration at C326 through amino acid mutation stabilizes the PKM2 tetramer and crystal structure further revealing the tetramer organization of PKM2-C326S. The PKM2-C326S mutant in cancer cells rewires glucose metabolism to mitochondrial respiration, significantly inhibiting tumor growth. In this work, we demonstrate that PKM2 sulfhydration by H2S inactivates PKM2 activity to promote tumorigenesis and inhibiting this process could be a potential therapeutic approach for targeting cancer metabolism.
Collapse
Grants
- National Science and Technology Council (Taiwan), 108-2314-B-007-003-MY3, 111-2320-B-007-005-MY3; National Tsing Hua University (NTHU), 111Q2713E1, 112Q2511E1, and 112Q2521E1, 113Q2524E1.
- National Science and Technology Council (Taiwan), 110-2320-B-007-004-MY3; National Health Research Institutes (Taiwan), NHRI-EX113-11124BI. National Tsing Hua University (NTHU), 112QI033E1
- National Science and Technology Council (Taiwan),110-2320-B-039-066; Ministry of Education (Taiwan), CMRC-CENTER-0
- National Science and Technology Council (Taiwan), 108-2311-B-007-002-MY3, 111-2311-B-007-009
Collapse
Affiliation(s)
- Rong-Hsuan Wang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Pin-Ru Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yue-Ting Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Chang Chen
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Yu-Hsin Chu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Chia-Chen Chien
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Po-Chen Chien
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Shao-Yun Lo
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Zhong-Liang Wang
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Min-Chen Tsou
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Ssu-Yu Chen
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Guang-Shen Chiu
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Ling Chen
- Institute of Molecular and Genomic Medicine, National Health Research Institutes, Zhunan, Taiwan
| | - Yi-Hsuan Wu
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Lily Hui-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Ching Wang
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Shu-Yi Lin
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Taiwan
| | - Hsing-Jien Kung
- College of Medical Science and Technology, PhD Program for Cancer Biology and Drug Discovery, Taipei Medical University, Taipei, Taiwan
| | - Lu-Hai Wang
- Chiese Medicine Research Center, and Institute of Integrated Medicine, China Medical University, Taichung City, Taiwan.
| | - Hui-Chun Cheng
- Institute of Bioinformatics and Structural Biology, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.
| | - Kai-Ti Lin
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
38
|
Atta S, Mandal A, Majumdar A. Generation of Thiosulfate, Selenite, Dithiosulfite, Perthionitrite, Nitric Oxide, and Reactive Chalcogen Species by Binuclear Zinc(II)-Chalcogenolato/-Polychalcogenido Complexes. Inorg Chem 2024; 63:15161-15176. [PMID: 39084849 DOI: 10.1021/acs.inorgchem.4c02527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
A comparative bioinspired reactivity study of new binuclear Zn(II) complexes featuring coordinated thiolate, selenolate, trisulfide and diselenide in relation with (i) the generation of reactive sulfur/selenium species (RSS/RSeS), (ii) the oxygen dependent oxidation and disproportionation of polysulfide (Sn2-) to produce sulfite (SO32-), thiosulfate (S2O32-) and sulfide (S2-) by sulfur oxygenase reductase (SOR), and (iii) the reaction of Sn2- with nitrite (NO2-) to generate thionitrite (SNO-), perthionitrite (SSNO-) and nitric oxide (NO), is presented. The binuclear Zn(II)-thiolate/selenolate complexes could react with elemental sulfur to generate RSS/RSeS while similar reactions involving elemental selenium could not generate RSeS. The dizinc(II)-S3 and the dizinc(II)-Se2 complexes could react with dioxygen (O2) to generate binuclear Zn(II) complexes featuring coordinated thiosulfate (S2O32-) and selenite (SeO32-), respectively. Finally, unlike the nonreactive nature of the dizinc(II)-Se2 complex toward NO2-, reaction of the dizinc(II)-S3 complex with NO2- produced a new binuclear Zn(II) complex featuring a coordinated dithiosulfite (S3O2-) along with the formation of perthionitrite (SSNO-), of which the latter subsequently produced nitric oxide (NO) and S42-. The present work, thus, demonstrates the comparative reactivity of a series of binuclear Zn(II)-chalcogenolato/-polychalcogenido complexes for the generation of S2O32-, SeO32-, S3O2-, SSNO-, NO and RSS/RSeS.
Collapse
Affiliation(s)
- Sayan Atta
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| | - Amit Mandal
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| | - Amit Majumdar
- School of Chemical Sciences, Indian Association for the Cultivation of Science, 2A & 2B Raja S. C. Mullick Road, Jadavpur, Kolkata 700032, West Bengal, India
| |
Collapse
|
39
|
Tang SM, Lu GZ, Lei XY, Yang XY, Tang GT, Yu J, Xie ZZ. Sodium thiosulfate: A donor or carrier signaling molecule for hydrogen sulfide? Nitric Oxide 2024; 149:67-74. [PMID: 38897561 DOI: 10.1016/j.niox.2024.06.004] [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: 03/03/2024] [Revised: 06/11/2024] [Accepted: 06/15/2024] [Indexed: 06/21/2024]
Abstract
Sodium thiosulfate has been used for decades in the treatment of calciphylaxis and cyanide detoxification, and has recently shown initial therapeutic promise in critical diseases such as neuronal ischemia, diabetes mellitus, heart failure and acute lung injury. However, the precise mechanism of sodium thiosulfate remains incompletely defined and sometimes contradictory. Although sodium thiosulfate has been widely accepted as a donor of hydrogen sulfide (H2S), emerging findings suggest that it is the executive signaling molecule for H2S and that its effects may not be dependent on H2S. This article presents an overview of the current understanding of sodium thiosulfate, including its synthesis, biological characteristics, and clinical applications of sodium thiosulfate, as well as the underlying mechanisms in vivo. We also discussed the interplay of sodium thiosulfate and H2S. Our review highlights sodium thiosulfate as a key player in sulfide signaling with the broad clinical potential for the future.
Collapse
Affiliation(s)
- Si-Miao Tang
- The Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, 421001, China
| | - Guo-Zhong Lu
- 922th Hospital of Hengyang, Hunan, 421001, China
| | - Xiao-Yong Lei
- The Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, 421001, China
| | - Xiao-Yan Yang
- The Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, 421001, China
| | - Guo-Tao Tang
- The Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, 421001, China
| | - Jia Yu
- The Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, 421001, China.
| | - Zhi-Zhong Xie
- The Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Institute of Pharmacy & Pharmacology, University of South China, Hengyang, Hunan, 421001, China.
| |
Collapse
|
40
|
Cobley JN, Margaritelis NV, Chatzinikolaou PN, Nikolaidis MG, Davison GW. Ten "Cheat Codes" for Measuring Oxidative Stress in Humans. Antioxidants (Basel) 2024; 13:877. [PMID: 39061945 PMCID: PMC11273696 DOI: 10.3390/antiox13070877] [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: 05/23/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Formidable and often seemingly insurmountable conceptual, technical, and methodological challenges hamper the measurement of oxidative stress in humans. For instance, fraught and flawed methods, such as the thiobarbituric acid reactive substances assay kits for lipid peroxidation, rate-limit progress. To advance translational redox research, we present ten comprehensive "cheat codes" for measuring oxidative stress in humans. The cheat codes include analytical approaches to assess reactive oxygen species, antioxidants, oxidative damage, and redox regulation. They provide essential conceptual, technical, and methodological information inclusive of curated "do" and "don't" guidelines. Given the biochemical complexity of oxidative stress, we present a research question-grounded decision tree guide for selecting the most appropriate cheat code(s) to implement in a prospective human experiment. Worked examples demonstrate the benefits of the decision tree-based cheat code selection tool. The ten cheat codes define an invaluable resource for measuring oxidative stress in humans.
Collapse
Affiliation(s)
- James N. Cobley
- The University of Dundee, Dundee DD1 4HN, UK
- Ulster University, Belfast BT15 1ED, Northern Ireland, UK;
| | - Nikos V. Margaritelis
- Aristotle University of Thessaloniki, 62122 Serres, Greece; (N.V.M.); (P.N.C.); (M.G.N.)
| | | | - Michalis G. Nikolaidis
- Aristotle University of Thessaloniki, 62122 Serres, Greece; (N.V.M.); (P.N.C.); (M.G.N.)
| | | |
Collapse
|
41
|
Stoltzfus AT, Ballot JG, Vignane T, Li H, Worth MM, Muller L, Siegler MA, Kane MA, Filipovic MR, Goldberg DP, Michel SLJ. Chemoselective Proteomics, Zinc Fingers, and a Zinc(II) Model for H 2S Mediated Persulfidation. Angew Chem Int Ed Engl 2024; 63:e202401003. [PMID: 38808693 PMCID: PMC11346292 DOI: 10.1002/anie.202401003] [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: 01/15/2024] [Indexed: 05/30/2024]
Abstract
The gasotransmitter hydrogen sulfide (H2S) is thought to be involved in the post-translational modification of cysteine residues to produce reactive persulfides. A persulfide-specific chemoselective proteomics approach with mammalian cells has identified a broad range of zinc finger (ZF) proteins as targets of persulfidation. Parallel studies with isolated ZFs show that persulfidation is mediated by ZnII, O2, and H2S, with intermediates involving oxygen- and sulfur-based radicals detected by mass spectrometry and optical spectroscopies. A small molecule ZnII complex exhibits analogous reactivity with H2S and O2, giving a persulfidated product. These data show that ZnII is not just a biological structural element, but also plays a critical role in mediating H2S-dependent persulfidation. ZF persulfidation appears to be a general post-translational modification and a possible conduit for H2S signaling. This work has implications for our understanding of H2S-mediated signaling and the regulation of ZFs in cellular physiology and development.
Collapse
Affiliation(s)
- Andrew T. Stoltzfus
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD, 21201, USA
| | - Jasper G. Ballot
- Department of Chemistry, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD, 21218, USA
| | - Thibaut Vignane
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V. Dortmund, Germany, 44139
| | - Haoju Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD, 21201, USA
| | - Madison M. Worth
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD, 21201, USA
| | - Ludovic Muller
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD, 21201, USA
| | - Maxime A. Siegler
- Department of Chemistry, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD, 21218, USA
| | - Maureen A. Kane
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD, 21201, USA
| | - Milos R. Filipovic
- Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V. Dortmund, Germany, 44139
| | - David P. Goldberg
- Department of Chemistry, Johns Hopkins University, 3400 N Charles Street, Baltimore, MD, 21218, USA
| | - Sarah L. J. Michel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, 20 Penn Street, Baltimore, MD, 21201, USA
| |
Collapse
|
42
|
Mann CG, MacArthur MR, Zhang J, Gong S, AbuSalim JE, Hunter CJ, Lu W, Agius T, Longchamp A, Allagnat F, Rabinowitz J, Mitchell JR, De Bock K, Mitchell SJ. Sulfur Amino Acid Restriction Enhances Exercise Capacity in Mice by Boosting Fat Oxidation in Muscle. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601041. [PMID: 39005372 PMCID: PMC11244859 DOI: 10.1101/2024.06.27.601041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Dietary restriction of the sulfur-containing amino acids methionine and cysteine (SAAR) improves body composition, enhances insulin sensitivity, and extends lifespan; benefits seen also with endurance exercise. Yet, the impact of SAAR on skeletal muscle remains largely unexplored. Here we demonstrate that one week of SAAR in sedentary, young, male mice increases endurance exercise capacity. Indirect calorimetry showed that SAAR increased lipid oxidation at rest and delayed the onset of carbohydrate utilization during exercise. Transcriptomic analysis revealed increased expression of genes involved in fatty acid catabolism especially in glycolytic muscle following SAAR. These findings were functionally supported by increased fatty acid circulatory turnover flux and muscle β-oxidation. Reducing lipid uptake from circulation through endothelial cell (EC)-specific CD36 deletion attenuated the running phenotype. Mechanistically, VEGF-signaling inhibition prevented exercise increases following SAAR, without affecting angiogenesis, implicating noncanonical VEGF signaling and EC CD36-dependent fatty acid transport in regulating exercise capacity by influencing muscle substrate availability.
Collapse
Affiliation(s)
- Charlotte G Mann
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Michael R MacArthur
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Jing Zhang
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Songlin Gong
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Jenna E AbuSalim
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Craig J. Hunter
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| | - Wenyun Lu
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Thomas Agius
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne 1005, Switzerland
| | - Alban Longchamp
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne 1005, Switzerland
- Transplant Center, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
- Center for Engineering in Medicine, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Florent Allagnat
- Department of Vascular Surgery, Lausanne University Hospital (CHUV), Lausanne 1005, Switzerland
| | - Joshua Rabinowitz
- Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
- Lewis-Sigler Institute of Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - James R Mitchell
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
- Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Katrien De Bock
- Department of Health Sciences and Technology, ETH Zurich, Zurich 8092, Switzerland
| | - Sarah J Mitchell
- Ludwig Institute for Cancer Research, Princeton University, Princeton, NJ 08544, USA
| |
Collapse
|
43
|
Miler M, Živanović J, Ajdžanović V, Milenkovic D, Cesar T, Filipović MR, Milošević V. Lemon extract reduces the hepatic oxidative stress and persulfidation levels by upregulating the Nrf2 and Trx1 expression in old rats. Biofactors 2024; 50:756-771. [PMID: 38194360 DOI: 10.1002/biof.2038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Accepted: 11/22/2023] [Indexed: 01/10/2024]
Abstract
Citrus flavanones are recognized as promising bioactives within the concept of healthy aging. Thus, the present study investigated the effects of a nutritionally relevant dose of lemon extract (LE) on liver redox regulation and persulfidation levels in 24-month-old Wistar rats. LE (40 mg/kg b.m.) was administered orally once daily for 4 weeks. Control groups received either vehicle (sunflower oil) or remained intact. The applied methodology considered qPCR, Western blot, protein persulfidation levels evaluation, histochemistry in line with immunofluorescence, liver biochemical assays (glutathione, total -SH groups and malonaldehyde; MDA), liver enzymes in serum and in silico analysis to explore the potential interaction/binding between the proteins studied in the paper. Our results showed that LE increased glutathione peroxidase (GPx), reductase (GR), glutamate-cysteine ligase catalytic and modifier subunit, respectively, as well as Nrf2 gene expressions, but decreased the expression of superoxide dismutase 2 (SOD2). Upon LE application, protein expression showed upregulation of NRF2, SOD2, GPx, GR, and thioredoxin 1 (Trx1). LE significantly decreased the protein persulfidation levels and concentration of MDA, a marker of oxidative damage in the cell. Histological analysis showed a normal liver histoarchitecture without pathological changes, aligning with the normal serum level of hepatic enzymes. Obtained results showed that LE, by modulating hepatic redox regulators Nrf2 and Trx1, diminishes oxidative stress and alters the persulfidation levels, suggesting a considerable beneficial antioxidant potential of lemon flavanones in the old-aged liver.
Collapse
Affiliation(s)
- Marko Miler
- Department of Cytology, Institute for Biological Research "Siniša Stanković" (IBISS)- National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Jasmina Živanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" (IBISS)- National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Vladimir Ajdžanović
- Department of Cytology, Institute for Biological Research "Siniša Stanković" (IBISS)- National Institute of the Republic of Serbia, University of Belgrade, Belgrade, Serbia
| | - Dragan Milenkovic
- Division of Cardiovascular Medicine, University of California Davis, Davis, California, USA
| | - Thais Cesar
- Graduate Program in Food, Nutrition and Food Engineering, Sao Paulo State University (UNESP), Araraquara, Brasil
| | - Miloš R Filipović
- Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V, Dortmund, Germany
| | - Verica Milošević
- Department of Anatomy, Faculty of Medicine, University of Niš, Niš, Serbia
| |
Collapse
|
44
|
Nishimura A, Tang X, Zhou L, Ito T, Kato Y, Nishida M. Sulfur metabolism as a new therapeutic target of heart failure. J Pharmacol Sci 2024; 155:75-83. [PMID: 38797536 DOI: 10.1016/j.jphs.2024.04.005] [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: 03/22/2024] [Revised: 04/07/2024] [Accepted: 04/21/2024] [Indexed: 05/29/2024] Open
Abstract
Sulfur-based redox signaling has long attracted attention as critical mechanisms underlying the development of cardiac diseases and resultant heart failure. Especially, post-translational modifications of cysteine (Cys) thiols in proteins mediate oxidative stress-dependent cardiac remodeling including myocardial hypertrophy, senescence, and interstitial fibrosis. However, we recently revealed the existence of Cys persulfides and Cys polysulfides in cells and tissues, which show higher redox activities than Cys and substantially contribute to redox signaling and energy metabolism. We have established simple evaluation methods that can detect polysulfides in proteins and inorganic polysulfides in cells and revealed that polysulfides abundantly expressed in normal hearts are dramatically catabolized by exposure to ischemic/hypoxic and environmental electrophilic stress, which causes vulnerability of the heart to mechanical load. Accumulation of hydrogen sulfide, a nucleophilic catabolite of persulfides/polysulfides, may lead to reductive stress in ischemic hearts, and perturbation of polysulfide catabolism can improve chronic heart failure after myocardial infarction in mice. This review focuses on the (patho)physiological role of sulfur metabolism in hearts, and proposes that sulfur catabolism during ischemic/hypoxic stress has great potential as a new therapeutic strategy for the treatment of ischemic heart failure.
Collapse
Affiliation(s)
- Akiyuki Nishimura
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems, NINS, Okazaki, 444-8787, Japan; SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan.
| | - Xiaokang Tang
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems, NINS, Okazaki, 444-8787, Japan; SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
| | - Liuchenzi Zhou
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems, NINS, Okazaki, 444-8787, Japan; SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan
| | - Tomoya Ito
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Yuri Kato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan
| | - Motohiro Nishida
- National Institute for Physiological Sciences, National Institutes of Natural Sciences (NINS), Okazaki, 444-8787, Japan; Exploratory Research Center on Life and Living Systems, NINS, Okazaki, 444-8787, Japan; SOKENDAI (The Graduate University for Advanced Studies), Okazaki, 444-8787, Japan; Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, 812-8582, Japan.
| |
Collapse
|
45
|
Kaleta K, Janik K, Rydz L, Wróbel M, Jurkowska H. Bridging the Gap in Cancer Research: Sulfur Metabolism of Leukemic Cells with a Focus on L-Cysteine Metabolism and Hydrogen Sulfide-Producing Enzymes. Biomolecules 2024; 14:746. [PMID: 39062461 PMCID: PMC11274876 DOI: 10.3390/biom14070746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/12/2024] [Accepted: 06/19/2024] [Indexed: 07/28/2024] Open
Abstract
Leukemias are cancers of the blood-forming system, representing a significant challenge in medical science. The development of leukemia cells involves substantial disturbances within the cellular machinery, offering hope in the search for effective selective treatments that could improve the 5-year survival rate. Consequently, the pathophysiological processes within leukemia cells are the focus of critical research. Enzymes such as cystathionine beta-synthase and sulfurtransferases like thiosulfate sulfurtransferase, 3-mercaptopyruvate sulfurtransferase, and cystathionine gamma-lyase play a vital role in cellular sulfur metabolism. These enzymes are essential to maintaining cellular homeostasis, providing robust antioxidant defenses, and supporting cell division. Numerous studies have demonstrated that cancerous processes can alter the expression and activity of these enzymes, uncovering potential vulnerabilities or molecular targets for cancer therapy. Recent laboratory research has indicated that certain leukemia cell lines may exhibit significant changes in the expression patterns of these enzymes. Analysis of the scientific literature and online datasets has confirmed variations in sulfur enzyme function in specific leukemic cell lines compared to normal leukocytes. This comprehensive review collects and analyzes available information on sulfur enzymes in normal and leukemic cell lines, providing valuable insights and identifying new research pathways in this field.
Collapse
Affiliation(s)
- Konrad Kaleta
- Students’ Scientific Group of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland;
| | - Klaudia Janik
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland; (K.J.); (L.R.); (M.W.)
| | - Leszek Rydz
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland; (K.J.); (L.R.); (M.W.)
| | - Maria Wróbel
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland; (K.J.); (L.R.); (M.W.)
| | - Halina Jurkowska
- Chair of Medical Biochemistry, Faculty of Medicine, Jagiellonian University Medical College, 7 Kopernika St., 31-034 Krakow, Poland; (K.J.); (L.R.); (M.W.)
| |
Collapse
|
46
|
He K, Zhang H, Tan B, Song C, Liang Z, Zhang L, Tian D, Xiao L, Xue H, Guo Q, Teng X, Jin S, An C, Wu Y. Hydrogen Sulfide Ameliorates Heart Aging by Downregulating Matrix Metalloproteinase-9. Cardiovasc Drugs Ther 2024:10.1007/s10557-024-07586-w. [PMID: 38884920 DOI: 10.1007/s10557-024-07586-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/21/2024] [Indexed: 06/18/2024]
Abstract
PURPOSE Aging contributes significantly to cardiovascular diseases and cardiac dysfunction, leading to the upregulation of matrix metalloproteinase-9 (MMP-9) in the heart and a significant decrease in hydrogen sulfide (H2S) content, coupled with impaired cardiac diastolic function. This study explores whether supplementing exogenous hydrogen sulfide during aging ameliorates the decline in H2S concentration in the heart, suppresses MMP-9 expression, and improves the age-associated impairment in cardiac morphology and function. METHODS We collected plasma from healthy individuals of different ages to determine the relationship between aging and H2S and MMP-9 levels through Elisa detection and liquid chromatography-tandem mass spectrometry (LC/MC) detection of plasma H2S content. Three-month-old mice were selected as the young group, while 18-month-old mice were selected as the old group, and sodium hydrosulfide (NaHS) was injected intraperitoneally from 15 months old until 18 months old as the old + NaHS group. Plasma MMP-9 content was detected using Elisa, plasma H2S content, cardiac H2S content, and cystathionine gamma-lyase (CSE) activity were detected using LC/MC, and cardiac function was detected using echocardiography. Heart structure was assessed using hematoxylin and eosin staining, Masone staining was used to detect the degree of cardiac fibrosis, while western blot was used to detect the expression of MMP-9, CSE, and aging marker proteins. Knockdown of MMP-9 and CSE in H9c2 cells using small interfering RNA was carried out to determine the upstream-downstream relationship between MMP-9 and CSE. RESULTS H2S content in the plasma of healthy individuals decreases with escalating age, whereas MMP-9 level rises with age progression. Aging leads to a decrease in H2S levels in the heart and plasma of mice, severe impairment of cardiac diastolic function, interstitial relaxation, and fibrosis of the heart. Supplementing with exogenous H2S can improve these phenomena. CONCLUSION H2S maintains the structure and function of the heart by inhibiting the expression of MMP-9 during the aging process.
Collapse
Affiliation(s)
- Kaichuan He
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Huaxing Zhang
- Core Facilities and Centers, Hebei Medical University, 050017, Hebei, China
| | - Bo Tan
- Clinical Pharmacokinetic Laboratory, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, 201203, Shanghai, China
| | - Chengqing Song
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Zihui Liang
- Clinical Practice Teaching Department, Hebei Medical University, 050017, Hebei, China
| | - Lixia Zhang
- Department of Medical Laboratory, Hebei Children's Hospital, 050017, Hebei, China
| | - Danyang Tian
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Lin Xiao
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Hongmei Xue
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Qi Guo
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Xu Teng
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Sheng Jin
- Department of Physiology, Hebei Medical University, 050017, Hebei, China
| | - Cuixia An
- Department of Psychiatry, the First Hospital of Hebei Medical University, 050031, Hebei, China.
| | - Yuming Wu
- Department of Physiology, Hebei Medical University, 050017, Hebei, China.
- Hebei Collaborative Innovation Center for Cardio-Cerebrovascular Disease, 050017, Hebei, China.
- Hebei Key Laboratory of Cardiovascular Homeostasis and Aging, 050017, Hebei, China.
- The Key Laboratory of Neural and Vascular Biology, Ministry of Education, 050017, Hebei, China.
| |
Collapse
|
47
|
Muñoz-Vargas MA, González-Gordo S, Aroca A, Romero LC, Gotor C, Palma JM, Corpas FJ. Persulfidome of Sweet Pepper Fruits during Ripening: The Case Study of Leucine Aminopeptidase That Is Positively Modulated by H 2S. Antioxidants (Basel) 2024; 13:719. [PMID: 38929158 PMCID: PMC11200738 DOI: 10.3390/antiox13060719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 06/06/2024] [Accepted: 06/11/2024] [Indexed: 06/28/2024] Open
Abstract
Protein persulfidation is a thiol-based oxidative posttranslational modification (oxiPTM) that involves the modification of susceptible cysteine thiol groups present in peptides and proteins through hydrogen sulfide (H2S), thus affecting their function. Using sweet pepper (Capsicum annuum L.) fruits as a model material at different stages of ripening (immature green and ripe red), endogenous persulfidated proteins (persulfidome) were labeled using the dimedone switch method and identified using liquid chromatography and mass spectrometry analysis (LC-MS/MS). A total of 891 persulfidated proteins were found in pepper fruits, either immature green or ripe red. Among these, 370 proteins were exclusively present in green pepper, 237 proteins were exclusively present in red pepper, and 284 proteins were shared between both stages of ripening. A comparative analysis of the pepper persulfidome with that described in Arabidopsis leaves allowed the identification of 25% of common proteins. Among these proteins, glutathione reductase (GR) and leucine aminopeptidase (LAP) were selected to evaluate the effect of persulfidation using an in vitro approach. GR activity was unaffected, whereas LAP activity increased by 3-fold after persulfidation. Furthermore, this effect was reverted through treatment with dithiothreitol (DTT). To our knowledge, this is the first persulfidome described in fruits, which opens new avenues to study H2S metabolism. Additionally, the results obtained lead us to hypothesize that LAP could be involved in glutathione (GSH) recycling in pepper fruits.
Collapse
Affiliation(s)
- María A. Muñoz-Vargas
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Stress, Development and Signaling in Plants, Estación Experimental del Zaidín Spanish National Research Council, CSIC, C/Profesor Albareda 1, 18008 Granada, Spain; (M.A.M.-V.); (S.G.-G.); (J.M.P.)
| | - Salvador González-Gordo
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Stress, Development and Signaling in Plants, Estación Experimental del Zaidín Spanish National Research Council, CSIC, C/Profesor Albareda 1, 18008 Granada, Spain; (M.A.M.-V.); (S.G.-G.); (J.M.P.)
| | - Angeles Aroca
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas and Universidad de Sevilla, Avenida Américo Vespucio 49, 41092 Sevilla, Spain; (A.A.); (L.C.R.); (C.G.)
| | - Luis C. Romero
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas and Universidad de Sevilla, Avenida Américo Vespucio 49, 41092 Sevilla, Spain; (A.A.); (L.C.R.); (C.G.)
| | - Cecilia Gotor
- Instituto de Bioquímica Vegetal y Fotosíntesis, Consejo Superior de Investigaciones Científicas and Universidad de Sevilla, Avenida Américo Vespucio 49, 41092 Sevilla, Spain; (A.A.); (L.C.R.); (C.G.)
| | - José M. Palma
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Stress, Development and Signaling in Plants, Estación Experimental del Zaidín Spanish National Research Council, CSIC, C/Profesor Albareda 1, 18008 Granada, Spain; (M.A.M.-V.); (S.G.-G.); (J.M.P.)
| | - Francisco J. Corpas
- Group of Antioxidants, Free Radicals and Nitric Oxide in Biotechnology, Food and Agriculture, Department of Stress, Development and Signaling in Plants, Estación Experimental del Zaidín Spanish National Research Council, CSIC, C/Profesor Albareda 1, 18008 Granada, Spain; (M.A.M.-V.); (S.G.-G.); (J.M.P.)
| |
Collapse
|
48
|
Li H, Stoltzfus AT, Michel SLJ. Mining proteomes for zinc finger persulfidation. RSC Chem Biol 2024; 5:572-585. [PMID: 38846077 PMCID: PMC11151867 DOI: 10.1039/d3cb00106g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 12/03/2023] [Indexed: 06/09/2024] Open
Abstract
Hydrogen sulfide (H2S) is an endogenous gasotransmitter that signals via persulfidation. There is evidence that the cysteine residues of certain zinc finger (ZF) proteins, a common type of cysteine rich protein, are modified to persulfides by H2S. To determine how frequently ZF persulfidation occurs in cells and identify the types of ZFs that are persulfidated, persulfide specific proteomics data were evaluated. 22 datasets from 16 studies were analyzed via a meta-analysis approach. Persulfidated ZFs were identified in a range of eukaryotic species, including Homo sapiens, Mus musculus, Rattus norvegicus, Arabidopsis thaliana, and Emiliania huxley (single-celled phytoplankton). The types of ZFs identified for each species encompassed all three common ZF ligand sets (4-cysteine, 3-cysteine-1-histidine, and 2-cysteine-2-hisitidine), indicating that persulfidation of ZFs is broad. Overlap analysis between different species identified several common ZFs. GO and KEGG analysis identified pathway enrichment for ubiquitin-dependent protein catabolic process and viral carcinogenesis. These collective findings support ZF persulfidation as a wide-ranging PTM that impacts all classes of ZFs.
Collapse
Affiliation(s)
- Haoju Li
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy Baltimore MD 21201 USA
| | - Andrew T Stoltzfus
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy Baltimore MD 21201 USA
| | - Sarah L J Michel
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy Baltimore MD 21201 USA
| |
Collapse
|
49
|
Amich J. The many roles of sulfur in the fungal-host interaction. Curr Opin Microbiol 2024; 79:102489. [PMID: 38754292 DOI: 10.1016/j.mib.2024.102489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/30/2024] [Accepted: 04/30/2024] [Indexed: 05/18/2024]
Abstract
Sulfur is an essential macronutrient for life, and consequently, all living organisms must acquire it from external sources to thrive and grow. Sulfur is a constituent of a multitude of crucial molecules, such as the S-containing proteinogenic amino acids cysteine and methionine; cofactors and prosthetic groups, such as coenzyme-A and iron-sulfur (Fe-S) clusters; and other essential organic molecules, such as glutathione or S-adenosylmethionine. Additionally, sulfur in cysteine thiols is an active redox group that plays paramount roles in protein stability, enzyme catalysis, and redox homeostasis. Furthermore, H2S is gaining more attention as a crucial signaling molecule that influences metabolism and physiological functions. Given its importance, it is not surprising that sulfur plays key roles in the host-pathogen interaction. However, in contrast to its well-recognized involvement in the plant-pathogen interaction, the specific contributions of sulfur to the human-fungal interaction are much less understood. In this short review, I highlight some of the most important known mechanisms and propose directions for further research.
Collapse
Affiliation(s)
- Jorge Amich
- Mycology Reference Laboratory (Laboratorio de Referencia e Investigación en Micología [LRIM]), National Centre for Microbiology, Instituto de Salud Carlos III (ISCIII), Majadahonda, Madrid, Spain.
| |
Collapse
|
50
|
Salti T, Braunstein I, Haimovich Y, Ziv T, Benhar M. Widespread S-persulfidation in activated macrophages as a protective mechanism against oxidative-inflammatory stress. Redox Biol 2024; 72:103125. [PMID: 38574432 PMCID: PMC11000178 DOI: 10.1016/j.redox.2024.103125] [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: 01/29/2024] [Revised: 03/10/2024] [Accepted: 03/12/2024] [Indexed: 04/06/2024] Open
Abstract
Acute inflammatory responses often involve the production of reactive oxygen and nitrogen species by innate immune cells, particularly macrophages. How activated macrophages protect themselves in the face of oxidative-inflammatory stress remains a long-standing question. Recent evidence implicates reactive sulfur species (RSS) in inflammatory responses; however, how endogenous RSS affect macrophage function and response to oxidative and inflammatory insults remains poorly understood. In this study, we investigated the endogenous pathways of RSS biogenesis and clearance in macrophages, with a particular focus on exploring how hydrogen sulfide (H2S)-mediated S-persulfidation influences macrophage responses to oxidative-inflammatory stress. We show that classical activation of mouse or human macrophages using lipopolysaccharide and interferon-γ (LPS/IFN-γ) triggers substantial production of H2S/RSS, leading to widespread protein persulfidation. Biochemical and proteomic analyses revealed that this surge in cellular S-persulfidation engaged ∼2% of total thiols and modified over 800 functionally diverse proteins. S-persulfidation was found to be largely dependent on the cystine importer xCT and the H2S-generating enzyme cystathionine γ-lyase and was independent of changes in the global proteome. We further investigated the role of the sulfide-oxidizing enzyme sulfide quinone oxidoreductase (SQOR), and found that it acts as a negative regulator of S-persulfidation. Elevated S-persulfidation following LPS/IFN-γ stimulation or SQOR inhibition was associated with increased resistance to oxidative stress. Upregulation of persulfides also inhibited the activation of the macrophage NLRP3 inflammasome and provided protection against inflammatory cell death. Collectively, our findings shed light on the metabolism and effects of RSS in macrophages and highlight the crucial role of persulfides in enabling macrophages to withstand and alleviate oxidative-inflammatory stress.
Collapse
Affiliation(s)
- Talal Salti
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Ilana Braunstein
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Yael Haimovich
- Smoler Proteomics Center and Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Tamar Ziv
- Smoler Proteomics Center and Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Moran Benhar
- Department of Biochemistry, Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|