1
|
Qian S, Chen G, Li R, Ma Y, Pan L, Wang X, Wang X. Disulfide stress and its role in cardiovascular diseases. Redox Biol 2024; 75:103297. [PMID: 39127015 PMCID: PMC11364009 DOI: 10.1016/j.redox.2024.103297] [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: 06/19/2024] [Revised: 07/30/2024] [Accepted: 07/31/2024] [Indexed: 08/12/2024] Open
Abstract
Cardiovascular disease (CVD) is one of the leading causes of mortality in humans, and oxidative stress plays a pivotal role in disease progression. This phenomenon typically arises from weakening of the cellular antioxidant system or excessive accumulation of peroxides. This review focuses on a specialized form of oxidative stress-disulfide stress-which is triggered by an imbalance in the glutaredoxin and thioredoxin antioxidant systems within the cell, leading to the accumulation of disulfide bonds. The genesis of disulfide stress is usually induced by extrinsic pathological factors that disrupt the thiol-dependent antioxidant system, manifesting as sustained glutathionylation of proteins, formation of abnormal intermolecular disulfide bonds between cysteine-rich proteins, or irreversible oxidation of thiol groups to sulfenic and sulfonic acids. Disulfide stress not only precipitates the collapse of the antioxidant system and the accumulation of reactive oxygen species, exacerbating oxidative stress, but may also initiate cellular inflammation, autophagy, and apoptosis through a cascade of signaling pathways. Furthermore, this review explores the detrimental effects of disulfide stress on the progression of various CVDs including atherosclerosis, hypertension, myocardial ischemia-reperfusion injury, diabetic cardiomyopathy, cardiac hypertrophy, and heart failure. This review also proposes several potential therapeutic avenues to improve the future treatment of CVDs.
Collapse
Affiliation(s)
- Shaoju Qian
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China; Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China; Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan, 453003, China
| | - Guanyu Chen
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Ruixue Li
- Department of Otolaryngology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, China
| | - Yinghua Ma
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Lin Pan
- School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, China
| | - Xiaoping Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China; Department of Human Anatomy and Histoembryology, Xinxiang Medical University, Xinxiang, China
| | - Xianwei Wang
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, China; Department of Human Anatomy and Histoembryology, Xinxiang Medical University, Xinxiang, China.
| |
Collapse
|
2
|
Mantle D, Dewsbury M, Hargreaves IP. The Ubiquinone-Ubiquinol Redox Cycle and Its Clinical Consequences: An Overview. Int J Mol Sci 2024; 25:6765. [PMID: 38928470 PMCID: PMC11203502 DOI: 10.3390/ijms25126765] [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: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 06/28/2024] Open
Abstract
Coenzyme Q10 (CoQ10) plays a key role in many aspects of cellular metabolism. For CoQ10 to function normally, continual interconversion between its oxidised (ubiquinone) and reduced (ubiquinol) forms is required. Given the central importance of this ubiquinone-ubiquinol redox cycle, this article reviews what is currently known about this process and the implications for clinical practice. In mitochondria, ubiquinone is reduced to ubiquinol by Complex I or II, Complex III (the Q cycle) re-oxidises ubiquinol to ubiquinone, and extra-mitochondrial oxidoreductase enzymes participate in the ubiquinone-ubiquinol redox cycle. In clinical terms, the outcome of deficiencies in various components associated with the ubiquinone-ubiquinol redox cycle is reviewed, with a particular focus on the potential clinical benefits of CoQ10 and selenium co-supplementation.
Collapse
Affiliation(s)
| | - Mollie Dewsbury
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK; (M.D.); (I.P.H.)
| | - Iain P. Hargreaves
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Liverpool L3 3AF, UK; (M.D.); (I.P.H.)
| |
Collapse
|
3
|
Yang B, Lin Y, Huang Y, Shen YQ, Chen Q. Thioredoxin (Trx): A redox target and modulator of cellular senescence and aging-related diseases. Redox Biol 2024; 70:103032. [PMID: 38232457 PMCID: PMC10827563 DOI: 10.1016/j.redox.2024.103032] [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: 10/14/2023] [Revised: 12/03/2023] [Accepted: 01/04/2024] [Indexed: 01/19/2024] Open
Abstract
Thioredoxin (Trx) is a compact redox-regulatory protein that modulates cellular redox state by reducing oxidized proteins. Trx exhibits dual functionality as an antioxidant and a cofactor for diverse enzymes and transcription factors, thereby exerting influence over their activity and function. Trx has emerged as a pivotal biomarker for various diseases, particularly those associated with oxidative stress, inflammation, and aging. Recent clinical investigations have underscored the significance of Trx in disease diagnosis, treatment, and mechanistic elucidation. Despite its paramount importance, the intricate interplay between Trx and cellular senescence-a condition characterized by irreversible growth arrest induced by multiple aging stimuli-remains inadequately understood. In this review, our objective is to present a comprehensive and up-to-date overview of the structure and function of Trx, its involvement in redox signaling pathways and cellular senescence, its association with aging and age-related diseases, as well as its potential as a therapeutic target. Our review aims to elucidate the novel and extensive role of Trx in senescence while highlighting its implications for aging and age-related diseases.
Collapse
Affiliation(s)
- Bowen Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yumeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Yibo Huang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Ying-Qiang Shen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China.
| | - Qianming Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Chinese Academy of Medical Sciences Research Unit of Oral Carcinogenesis and Management, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
4
|
Shcholok T, Eftekharpour E. Insights into the Multifaceted Roles of Thioredoxin-1 System: Exploring Knockout Murine Models. BIOLOGY 2024; 13:180. [PMID: 38534450 DOI: 10.3390/biology13030180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 03/05/2024] [Accepted: 03/07/2024] [Indexed: 03/28/2024]
Abstract
Redox balance is increasingly identified as a major player in cellular signaling. A fundamentally simple reaction of oxidation and reduction of cysteine residues in cellular proteins is the central concept in this complex regulatory mode of protein function. Oxidation of key cysteine residues occurs at the physiological levels of reactive oxygen species (ROS), but they are reduced by a supply of thiol antioxidant molecules including glutathione, glutaredoxin, and thioredoxin. While these molecules show complex compensatory roles in experimental conditions, transgenic animal models provide a comprehensive picture to pinpoint the role of each antioxidant. In this review, we have specifically focused on the available literature on thioredoxin-1 system transgenic models that include thioredoxin and thioredoxin reductase proteins. As the identification of thioredoxin protein targets is technically challenging, the true contribution of this system in maintaining cellular balance remains unidentified, including the role of this system in the brain.
Collapse
Affiliation(s)
- Tetiana Shcholok
- Department of Physiology and Pathophysiology, University of Manitoba, 631-BMSB, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada
| | - Eftekhar Eftekharpour
- Department of Physiology and Pathophysiology, University of Manitoba, 631-BMSB, 745 Bannatyne Ave, Winnipeg, MB R3E 0J9, Canada
| |
Collapse
|
5
|
Angelone T, Rocca C, Lionetti V, Penna C, Pagliaro P. Expanding the Frontiers of Guardian Antioxidant Selenoproteins in Cardiovascular Pathophysiology. Antioxid Redox Signal 2024; 40:369-432. [PMID: 38299513 DOI: 10.1089/ars.2023.0285] [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] [Indexed: 02/02/2024]
Abstract
Significance: Physiological levels of reactive oxygen and nitrogen species (ROS/RNS) function as fundamental messengers for many cellular and developmental processes in the cardiovascular system. ROS/RNS involved in cardiac redox-signaling originate from diverse sources, and their levels are tightly controlled by key endogenous antioxidant systems that counteract their accumulation. However, dysregulated redox-stress resulting from inefficient removal of ROS/RNS leads to inflammation, mitochondrial dysfunction, and cell death, contributing to the development and progression of cardiovascular disease (CVD). Recent Advances: Basic and clinical studies demonstrate the critical role of selenium (Se) and selenoproteins (unique proteins that incorporate Se into their active site in the form of the 21st proteinogenic amino acid selenocysteine [Sec]), including glutathione peroxidase and thioredoxin reductase, in cardiovascular redox homeostasis, representing a first-line enzymatic antioxidant defense of the heart. Increasing attention has been paid to emerging selenoproteins in the endoplasmic reticulum (ER) (i.e., a multifunctional intracellular organelle whose disruption triggers cardiac inflammation and oxidative stress, leading to multiple CVD), which are crucially involved in redox balance, antioxidant activity, and calcium and ER homeostasis. Critical Issues: This review focuses on endogenous antioxidant strategies with therapeutic potential, particularly selenoproteins, which are very promising but deserve more detailed and clinical studies. Future Directions: The importance of selective selenoproteins in embryonic development and the consequences of their mutations and inborn errors highlight the need to improve knowledge of their biological function in myocardial redox signaling. This could facilitate the development of personalized approaches for the diagnosis, prevention, and treatment of CVD. Antioxid. Redox Signal. 40, 369-432.
Collapse
Affiliation(s)
- Tommaso Angelone
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Rende, Italy
- National Institute of Cardiovascular Research (INRC), Bologna, Italy
| | - Carmine Rocca
- Cellular and Molecular Cardiovascular Pathophysiology Laboratory, Department of Biology, Ecology and Earth Sciences (DiBEST), University of Calabria, Rende, Italy
| | - Vincenzo Lionetti
- Unit of Translational Critical Care Medicine, Laboratory of Basic and Applied Medical Sciences, Interdisciplinary Research Center "Health Science," Scuola Superiore Sant'Anna, Pisa, Italy
- UOSVD Anesthesiology and Intensive Care Medicine, Fondazione Toscana "Gabriele Monasterio," Pisa, Italy
| | - Claudia Penna
- National Institute of Cardiovascular Research (INRC), Bologna, Italy
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| | - Pasquale Pagliaro
- National Institute of Cardiovascular Research (INRC), Bologna, Italy
- Department of Clinical and Biological Sciences, University of Turin, Orbassano, Italy
| |
Collapse
|
6
|
Perez V, Zaobornyj T, Vico T, Vanasco V, Marchini T, Godoy E, Alvarez S, Evelson P, Donato M, Gelpi RJ, D'Annunzio V. Middle-age abolishes cardioprotection conferred by thioredoxin-1 in mice. Arch Biochem Biophys 2024; 753:109880. [PMID: 38171410 DOI: 10.1016/j.abb.2023.109880] [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: 11/06/2023] [Revised: 12/29/2023] [Accepted: 12/30/2023] [Indexed: 01/05/2024]
Abstract
Thioredoxin-1 (Trx1) has cardioprotective effects on ischemia/reperfusion (I/R) injury, although its role in ischemic postconditioning (PostC) in middle-aged mice is not understood. This study aimed to evaluate if combining two cardioprotective strategies, such as Trx1 overexpression and PostC, could exert a synergistic effect in reducing infarct size in middle-aged mice. Young or middle-aged wild-type mice (Wt), transgenic mice overexpressing Trx1, and dominant negative (DN-Trx1) mutant of Trx1 mice were used. Mice hearts were subjected to I/R or PostC protocol. Infarct size, hydrogen peroxide (H2O2) production, protein nitration, Trx1 activity, mitochondrial function, and Trx1, pAkt and pGSK3β expression were measured. PostC could not reduce infarct size even in the presence of Trx1 overexpression in middle-aged mice. This finding was accompanied by a lack of Akt and GSK3β phosphorylation, and Trx1 expression (in Wt group). Trx1 activity was diminished and H2O2 production and protein nitration were increased in middle-age. The respiratory control rate dropped after I/R in Wt-Young and PostC restored this value, but not in middle-aged groups. Our results showed that Trx1 plays a key role in the PostC protection mechanism in young but not middle-aged mice, even in the presence of Trx1 overexpression.
Collapse
Affiliation(s)
- V Perez
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Patología - Institute of Cardiovascular Physiopathology, Argentina
| | - T Zaobornyj
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET), Argentina
| | - T Vico
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET), Argentina
| | - V Vanasco
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET), Argentina
| | - T Marchini
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET), Argentina
| | - E Godoy
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Patología - Institute of Cardiovascular Physiopathology, Argentina
| | - S Alvarez
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET), Argentina
| | - P Evelson
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET), Argentina
| | - M Donato
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Patología - Institute of Cardiovascular Physiopathology, Argentina
| | - R J Gelpi
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Patología - Institute of Cardiovascular Physiopathology, Argentina
| | - V D'Annunzio
- Universidad de Buenos Aires, Facultad de Ciencias Médicas, Departamento de Patología - Institute of Cardiovascular Physiopathology, Argentina.
| |
Collapse
|
7
|
Nagarajan N, Oka SI, Nah J, Wu C, Zhai P, Mukai R, Xu X, Kashyap S, Huang CY, Sung EA, Mizushima W, Titus AS, Takayama K, Mourad Y, Francisco J, Liu T, Chen T, Li H, Sadoshima J. Thioredoxin 1 promotes autophagy through transnitrosylation of Atg7 during myocardial ischemia. J Clin Invest 2023; 133:e162326. [PMID: 36480290 PMCID: PMC9888389 DOI: 10.1172/jci162326] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 12/07/2022] [Indexed: 12/13/2022] Open
Abstract
Modification of cysteine residues by oxidative and nitrosative stress affects structure and function of proteins, thereby contributing to the pathogenesis of cardiovascular disease. Although the major function of thioredoxin 1 (Trx1) is to reduce disulfide bonds, it can also act as either a denitrosylase or transnitrosylase in a context-dependent manner. Here we show that Trx1 transnitrosylates Atg7, an E1-like enzyme, thereby stimulating autophagy. During ischemia, Trx1 was oxidized at Cys32-Cys35 of the oxidoreductase catalytic center and S-nitrosylated at Cys73. Unexpectedly, Atg7 Cys545-Cys548 reduced the disulfide bond in Trx1 at Cys32-Cys35 through thiol-disulfide exchange and this then allowed NO to be released from Cys73 in Trx1 and transferred to Atg7 at Cys402. Experiments conducted with Atg7 C402S-knockin mice showed that S-nitrosylation of Atg7 at Cys402 promotes autophagy by stimulating E1-like activity, thereby protecting the heart against ischemia. These results suggest that the thiol-disulfide exchange and the NO transfer are functionally coupled, allowing oxidized Trx1 to mediate a salutary effect during myocardial ischemia through transnitrosylation of Atg7 and stimulation of autophagy.
Collapse
Affiliation(s)
- Narayani Nagarajan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Shin-ichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jihoon Nah
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Changgong Wu
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Peiyong Zhai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Risa Mukai
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Xiaoyong Xu
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Department of Cardiology, Ningbo Medical Center Lihuili Hospital, Ningbo, Zhejiang, China
| | - Sanchita Kashyap
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Chun-Yang Huang
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
- Division of Cardiovascular Surgery, Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
- Institute of Clinical Medicine, School of Medicine National Yang-Ming University, Taipei, Taiwan
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Wataru Mizushima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Allen Sam Titus
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Koichiro Takayama
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Youssef Mourad
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Jamie Francisco
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| | - Tong Liu
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Tong Chen
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Hong Li
- Center for Advanced Proteomics Research, Department of Microbiology, Biochemistry, and Molecular Genetics, Rutgers New Jersey Medical School and Cancer Institute of New Jersey, Newark, New Jersey, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, Newark, New Jersey, USA
| |
Collapse
|
8
|
Dagdeviren S, Lee RT, Wu N. Physiological and Pathophysiological Roles of Thioredoxin Interacting Protein: A Perspective on Redox Inflammation and Metabolism. Antioxid Redox Signal 2023; 38:442-460. [PMID: 35754346 PMCID: PMC9968628 DOI: 10.1089/ars.2022.0022] [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: 02/25/2022] [Accepted: 06/12/2022] [Indexed: 11/12/2022]
Abstract
Significance: Thioredoxin interacting protein (TXNIP) is a member of the arrestin fold superfamily with important cellular functions, including cellular transport, mitochondrial energy generation, and protein cycling. It is the only arrestin-domain protein known to covalently bind to thioredoxin and plays roles in glucose metabolism, inflammation, apoptosis, and cancer. Recent Advances: The crystal structure of the TXNIP-thioredoxin complex provided details about this fascinating interaction. Recent studies showed that TXNIP is induced by endoplasmic reticulum (ER) stress, activates NLR family pyrin domain containing 3 (NLRP3) inflammasomes, and can regulate glucose transport into cells. The tumor suppressor role of TXNIP in various cancer types and the role of TXNIP in fructose absorption are now described. Critical Issues: The influence of TXNIP on redox state is more complex than its interaction with thioredoxin. Future Directions: It is incompletely understood which functions of TXNIP are thioredoxin-dependent. It is also unclear whether TXNIP binding can inhibit glucose transporters without endocytosis. TXNIP-regulated control of ER stress should also be investigated further. Antioxid. Redox Signal. 38, 442-460.
Collapse
Affiliation(s)
- Sezin Dagdeviren
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Richard T. Lee
- Department of Stem Cell and Regenerative Biology and the Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, USA
| | - Ning Wu
- Van Andel Institute, Grand Rapids, Michigan, USA
| |
Collapse
|
9
|
Yu Y, Wu T, Lu Y, Zhao W, Zhang J, Chen Q, Ge G, Hua Y, Chen K, Ullah I, Zhang F. Exosomal thioredoxin-1 from hypoxic human umbilical cord mesenchymal stem cells inhibits ferroptosis in doxorubicin-induced cardiotoxicity via mTORC1 signaling. Free Radic Biol Med 2022; 193:108-121. [PMID: 36241072 DOI: 10.1016/j.freeradbiomed.2022.10.268] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 12/13/2022]
Abstract
Doxorubicin (DOX), a clinical chemotherapeutic drug, is often annoyed by its cardiotoxicity which involves ferroptosis in its pathological progress. Human umbilical cord mesenchymal stem cells (HucMSCs)-derived exosomes (HucMSCs-Exo) are proven effective in treating cardiovascular diseases. This study aimed to compare the therapeutic effects between normoxic HucMSCs-Exo (Exo) and hypoxic HucMSCs-Exo (Hypo-Exo) on DOX-induced ferroptosis and explore the underlying mechanisms. An acute cardiotoxicity model was successfully constructed by administrating two doses intraperitoneal injections of DOX (25 mg/kg in total). Exo and Hypo-Exo were extracted by ultracentrifugation and characterized. Compared with Exo, Hypo-Exo and Ferrostatin-1 (Fer-1) exerted superior effects on inhibiting DOX-induced ferroptosis, as evidenced by decreasing malondialdehyde (MDA), iron content and increasing glutathione (GSH) level as well as ferroptosis-related genes expression including prostaglandin-endoperoxide synthase 2 (Ptgs2) mRNA level and glutathione peroxidase 4 (GPX4) protein level. Based on quantitative proteomics analysis, we found that thioredoxin1 (Trx1) was remarkably upregulated in Hypo-Exo and exhibited anti-ferroptosis activity via activating the mechanistic target of rapamycin complex 1 (mTORC1) in neonatal rat cardiomyocytes (NRCMs). Trx1 knockdown and rapamycin (an mTORC1 inhibitor) partially abolished the protective effects of Hypo-Exo. Furthermore, our data indicated that solute carrier family 7 member 11 (SLC7A11) was critical for GPX4 protein synthesis. In conclusion, Hypo-Exo exhibited a better suppression of ferroptosis in DOX-induced cardiotoxicity. Trx1-mediated mTORC1 activation is critical for the Hypo-Exo anti-ferroptosis process, which involves increased GPX4 protein synthesis and decreased iron overload. This study indicated that Hypo-Exo may present a potential strategy against ferroptosis in DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Yue Yu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Tianyu Wu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yao Lu
- Department of Cardiology, Xuzhou Central Hospital, The Affiliated XuZhou Hospital of Nanjing Medical University, Xuzhou, 221009, Jiangsu, China
| | - Wei Zhao
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Jian Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Qiushi Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Gaoyuan Ge
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Yan Hua
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Kaiyan Chen
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Inam Ullah
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China
| | - Fengxiang Zhang
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, Jiangsu, China.
| |
Collapse
|
10
|
Thioredoxin deficiency increases oxidative stress and causes bilateral symmetrical degeneration in rat midbrain. Neurobiol Dis 2022; 175:105921. [DOI: 10.1016/j.nbd.2022.105921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 10/26/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022] Open
|
11
|
Carew NT, Schmidt HM, Yuan S, Galley JC, Hall R, Altmann HM, Hahn SA, Miller MP, Wood KC, Gabris B, Stapleton MC, Hartwick S, Fazzari M, Wu YL, Trebak M, Kaufman BA, McTiernan CF, Schopfer FJ, Navas P, Thibodeau PH, McNamara DM, Salama G, Straub AC. Loss of cardiomyocyte CYB5R3 impairs redox equilibrium and causes sudden cardiac death. J Clin Invest 2022; 132:e147120. [PMID: 36106636 PMCID: PMC9479700 DOI: 10.1172/jci147120] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Accepted: 07/19/2022] [Indexed: 01/04/2023] Open
Abstract
Sudden cardiac death (SCD) in patients with heart failure (HF) is allied with an imbalance in reduction and oxidation (redox) signaling in cardiomyocytes; however, the basic pathways and mechanisms governing redox homeostasis in cardiomyocytes are not fully understood. Here, we show that cytochrome b5 reductase 3 (CYB5R3), an enzyme known to regulate redox signaling in erythrocytes and vascular cells, is essential for cardiomyocyte function. Using a conditional cardiomyocyte-specific CYB5R3-knockout mouse, we discovered that deletion of CYB5R3 in male, but not female, adult cardiomyocytes causes cardiac hypertrophy, bradycardia, and SCD. The increase in SCD in CYB5R3-KO mice is associated with calcium mishandling, ventricular fibrillation, and cardiomyocyte hypertrophy. Molecular studies reveal that CYB5R3-KO hearts display decreased adenosine triphosphate (ATP), increased oxidative stress, suppressed coenzyme Q levels, and hemoprotein dysregulation. Finally, from a translational perspective, we reveal that the high-frequency missense genetic variant rs1800457, which translates into a CYB5R3 T117S partial loss-of-function protein, associates with decreased event-free survival (~20%) in Black persons with HF with reduced ejection fraction (HFrEF). Together, these studies reveal a crucial role for CYB5R3 in cardiomyocyte redox biology and identify a genetic biomarker for persons of African ancestry that may potentially increase the risk of death from HFrEF.
Collapse
Affiliation(s)
- Nolan T. Carew
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | - Heidi M. Schmidt
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | - Shuai Yuan
- Heart, Lung, Blood and Vascular Medicine Institute
| | - Joseph C. Galley
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | - Robert Hall
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | | | | | | | - Katherine C. Wood
- Heart, Lung, Blood and Vascular Medicine Institute
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, and
| | - Bethann Gabris
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Margaret C. Stapleton
- Department of Developmental Biology and Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sean Hartwick
- Department of Developmental Biology and Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | | | - Yijen L. Wu
- Department of Developmental Biology and Rangos Research Center Animal Imaging Core, Children’s Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mohamed Trebak
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | - Brett A. Kaufman
- Heart, Lung, Blood and Vascular Medicine Institute
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Charles F. McTiernan
- Heart, Lung, Blood and Vascular Medicine Institute
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Francisco J. Schopfer
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
| | - Placido Navas
- Andalusian Center for Developmental Biology and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Universidad Pablo de Olavide-CSIC-JA, Sevilla, Spain
| | | | - Dennis M. McNamara
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Guy Salama
- Heart, Lung, Blood and Vascular Medicine Institute
- Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Adam C. Straub
- Heart, Lung, Blood and Vascular Medicine Institute
- Department of Pharmacology and Chemical Biology
- Center for Microvascular Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
12
|
Matsuo I, Kawamura N, Ohnuki Y, Suita K, Ishikawa M, Matsubara T, Mototani Y, Ito A, Hayakawa Y, Nariyama M, Morii A, Kiyomoto K, Tsunoda M, Gomi K, Okumura S. Role of TLR4 signaling on Porphyromonas gingivalis LPS-induced cardiac dysfunction in mice. PLoS One 2022; 17:e0258823. [PMID: 35648750 PMCID: PMC9159598 DOI: 10.1371/journal.pone.0258823] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 05/14/2022] [Indexed: 12/02/2022] Open
Abstract
Oral infections, particularly periodontitis, are a well-established risk factor for cardiovascular diseases, although the molecular mechanisms involved remain elusive. The aims of the present study were to investigate the effects of lipopolysaccharide derived from Porphyromonas gingivalis (PG-LPS) on cardiac function in mice, and to elucidate the underlying mechanisms. Mice (C57BL/6) were injected with PG-LPS (0.8 mg/kg/day) with or without an inhibitor of Toll-like receptor 4 (TLR4) signaling (TAK-242, 0.8 mg/kg/day) for 4 weeks. Left ventricular ejection function was significantly decreased at 1 week (from 67 ± 0.5 to 58 ± 1.2%) and remained low at 4 weeks (57 ± 1.0%). The number of apoptotic myocytes was increased (approximately 7.4-fold), the area of fibrosis was increased (approximately 3.3-fold) and the number of 8-hydroxydeoxyguanosine-positive myocytes, a sensitive indicator of oxidative DNA damage, was increased (approximately 7.6-fold) at 4 weeks in the heart of PG-LPS treated mice. However, levels of various serum pro-inflammatory cytokines in PG-LPS-treated mice were similar to those in control mice. The impairment of cardiac function in PG-LPS-treated mice appears to involve activation of TLR4-NADPH oxidase (NOX) 4 signaling, leading to abundant production of reactive oxygen species and Ca2+ leakage from sarcoplastic reticulumn induced by calmodulin kinase II (CaMKII)-mediated phosphorylation of phospholamban (at Thr-17) and ryanodine receptor 2 (at Ser-2448). Pharmacological inhibition of TLR4 with TAK-242 attenuated the changes in cardiac function in PG-LPS-treated mice. Our results indicate that TLR4-NOX4 signaling may be a new therapeutic target for treatment of cardiovascular diseases in patients with periodontitis.
Collapse
Affiliation(s)
- Ichiro Matsuo
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Naoya Kawamura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Takehiro Matsubara
- Division of BioBank, Center for Comprehensive Genomic Medicine, Okayama University Hospital, Okayama, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshio Hayakawa
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Akinaka Morii
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenichi Kiyomoto
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Michinori Tsunoda
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kazuhiro Gomi
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- * E-mail:
| |
Collapse
|
13
|
The Role and Mechanism of Essential Selenoproteins for Homeostasis. Antioxidants (Basel) 2022; 11:antiox11050973. [PMID: 35624837 PMCID: PMC9138076 DOI: 10.3390/antiox11050973] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 05/05/2022] [Accepted: 05/10/2022] [Indexed: 12/25/2022] Open
Abstract
Selenium (Se) is one of the essential trace elements that plays a biological role in the body, mainly in the form of selenoproteins. Selenoproteins can be involved in the regulation of oxidative stress, endoplasmic reticulum (ER) stress, antioxidant defense, immune and inflammatory responses and other biological processes, including antioxidant, anti-inflammation, anti-apoptosis, the regulation of immune response and other functions. Over-loading or lack of Se causes certain damage to the body. Se deficiency can reduce the expression and activity of selenoproteins, disrupt the normal physiological function of cells and affect the body in antioxidant, immunity, toxin antagonism, signaling pathways and other aspects, thus causing different degrees of damage to the body. Se intake is mainly in the form of dietary supplements. Due to the important role of Se, people pay increasingly more attention to Se-enriched foods, which also lays a foundation for better research on the mechanism of selenoproteins in the future. In this paper, the synthesis and mechanism of selenoproteins, as well as the role and mechanism of selenoproteins in the regulation of diseases, are reviewed. Meanwhile, the future development of Se-enriched products is prospected, which is of great significance to further understand the role of Se.
Collapse
|
14
|
The Impact of Selenium Deficiency on Cardiovascular Function. Int J Mol Sci 2021; 22:ijms221910713. [PMID: 34639053 PMCID: PMC8509311 DOI: 10.3390/ijms221910713] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 09/28/2021] [Accepted: 09/29/2021] [Indexed: 01/04/2023] Open
Abstract
Selenium (Se) is an essential trace element that is necessary for various metabolic processes, including protection against oxidative stress, and proper cardiovascular function. The role of Se in cardiovascular health is generally agreed upon to be essential yet not much has been defined in terms of specific functions. Se deficiency was first associated with Keshan’s Disease, an endemic disease characterized by cardiomyopathy and heart failure. Since then, Se deficiency has been associated with multiple cardiovascular diseases, including myocardial infarction, heart failure, coronary heart disease, and atherosclerosis. Se, through its incorporation into selenoproteins, is vital to maintain optimal cardiovascular health, as selenoproteins are involved in numerous crucial processes, including oxidative stress, redox regulation, thyroid hormone metabolism, and calcium flux, and inadequate Se may disrupt these processes. The present review aims to highlight the importance of Se in cardiovascular health, provide updated information on specific selenoproteins that are prominent for proper cardiovascular function, including how these proteins interact with microRNAs, and discuss the possibility of Se as a potential complemental therapy for prevention or treatment of cardiovascular disease.
Collapse
|
15
|
Chelko SP, Keceli G, Carpi A, Doti N, Agrimi J, Asimaki A, Beti CB, Miyamoto M, Amat-Codina N, Bedja D, Wei AC, Murray B, Tichnell C, Kwon C, Calkins H, James CA, O'Rourke B, Halushka MK, Melloni E, Saffitz JE, Judge DP, Ruvo M, Kitsis RN, Andersen P, Di Lisa F, Paolocci N. Exercise triggers CAPN1-mediated AIF truncation, inducing myocyte cell death in arrhythmogenic cardiomyopathy. Sci Transl Med 2021; 13:13/581/eabf0891. [PMID: 33597260 DOI: 10.1126/scitranslmed.abf0891] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 01/27/2021] [Indexed: 12/15/2022]
Abstract
Myocyte death occurs in many inherited and acquired cardiomyopathies, including arrhythmogenic cardiomyopathy (ACM), a genetic heart disease plagued by the prevalence of sudden cardiac death. Individuals with ACM and harboring pathogenic desmosomal variants, such as desmoglein-2 (DSG2), often show myocyte necrosis with progression to exercise-associated heart failure. Here, we showed that homozygous Dsg2 mutant mice (Dsg2 mut/mut), a model of ACM, die prematurely during swimming and display myocardial dysfunction and necrosis. We detected calcium (Ca2+) overload in Dsg2 mut/mut hearts, which induced calpain-1 (CAPN1) activation, association of CAPN1 with mitochondria, and CAPN1-induced cleavage of mitochondrial-bound apoptosis-inducing factor (AIF). Cleaved AIF translocated to the myocyte nucleus triggering large-scale DNA fragmentation and cell death, an effect potentiated by mitochondrial-driven AIF oxidation. Posttranslational oxidation of AIF cysteine residues was due, in part, to a depleted mitochondrial thioredoxin-2 redox system. Hearts from exercised Dsg2 mut/mut mice were depleted of calpastatin (CAST), an endogenous CAPN1 inhibitor, and overexpressing CAST in myocytes protected against Ca2+ overload-induced necrosis. When cardiomyocytes differentiated from Dsg2 mut/mut embryonic stem cells (ES-CMs) were challenged with β-adrenergic stimulation, CAPN1 inhibition attenuated CAPN1-induced AIF truncation. In addition, pretreatment of Dsg2 mut/mut ES-CMs with an AIF-mimetic peptide, mirroring the cyclophilin-A (PPIA) binding site of AIF, blocked PPIA-mediated AIF-nuclear translocation, and reduced both apoptosis and necrosis. Thus, preventing CAPN1-induced AIF-truncation or barring binding of AIF to the nuclear chaperone, PPIA, may avert myocyte death and, ultimately, disease progression to heart failure in ACM and likely other forms of cardiomyopathies.
Collapse
Affiliation(s)
- Stephen P Chelko
- Department of Biomedical Sciences, Florida State University, Tallahassee, FL 32306, USA. .,Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Gizem Keceli
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Andrea Carpi
- Department of Biomedical Sciences, University of Padova, Padova 35122, Italy
| | - Nunzianna Doti
- Institute of Biostructures and Bioimaging, CNR, Naples 80134, Italy
| | - Jacopo Agrimi
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Angeliki Asimaki
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London WC1E 6BS, UK
| | - Carlos Bueno Beti
- Molecular and Clinical Sciences Research Institute, St. George's, University of London, London WC1E 6BS, UK
| | - Matthew Miyamoto
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Nuria Amat-Codina
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Djahida Bedja
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.,Australian School of Advanced Medicine, Macquarie University, Sydney, NSW 2109, Australia
| | - An-Chi Wei
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Brittney Murray
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Crystal Tichnell
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Chulan Kwon
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Hugh Calkins
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Cynthia A James
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Brian O'Rourke
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Marc K Halushka
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Edon Melloni
- Department of Medicine, University of Genova, Genova 16126, Italy
| | - Jeffrey E Saffitz
- Department of Pathology, Beth Israel Deaconess Medical Center, Boston, MA 20115, USA
| | - Daniel P Judge
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA.,Medical University of South Carolina, Charleston, SC 29425, USA
| | - Menotti Ruvo
- Institute of Biostructures and Bioimaging, CNR, Naples 80134, Italy
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Peter Andersen
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Padova 35122, Italy
| | - Nazareno Paolocci
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21205, USA. .,Department of Biomedical Sciences, University of Padova, Padova 35122, Italy
| |
Collapse
|
16
|
dos Santos PP, Rafacho BPM, Gonçalves AF, Pires VCM, Roscani MG, Azevedo PS, Polegato BF, Minicucci MF, Fernandes AAH, Tanni SE, Zornoff LAM, de Paiva SAR. Vitamin D Supplementation Induces Cardiac Remodeling in Rats: Association with Thioredoxin-Interacting Protein and Thioredoxin. Arq Bras Cardiol 2021; 116:970-978. [PMID: 34008824 PMCID: PMC8121481 DOI: 10.36660/abc.20190633] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2019] [Revised: 04/14/2020] [Accepted: 06/10/2020] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Vitamin D (VD) has been shown to play an important role in cardiac function. However, this vitamin exerts a biphasic "dose response" curve in cardiovascular pathophysiology and may cause deleterious effects, even in non-toxic doses. VD exerts its cellular functions by binding to VD receptor. Additionally, it was identified that the thioredoxin-interacting protein (TXNIP) expression is positively regulated by VD. TXNIP modulate different cell signaling pathways that may be important for cardiac remodeling. OBJECTIVE To evaluate whether VD supplementation lead to cardiac remodeling and if TXNIP and thioredoxin (Trx) proteins are associated with the process. METHODS A total of 250 Male Wistar rats were allocated into three groups: control (C, n=21), with no VD supplementation; VD3 (n = 22) and VD10 (n=21), supplemented with 3,000 and 10,000 IU of VD/ kg of chow respectively, for two months. The groups were compared by one-way analysis of variance (ANOVA) and Holm-Sidak post hoc analysis, (variables with normal distribution), or by Kruskal-Wallis test and Dunn's test post hoc analysis. The significance level for all tests was 5%. RESULTS TXNIP protein expression was higher and Trx activity was lower in VD10. The animals supplemented with VD showed increased lipid hydroperoxide and decreased superoxide dismutase and glutathione peroxidase. The protein Bcl-2 was lower in VD10. There was a decrease in fatty acid β-oxidation, tricarboxylic acid cycle and electron transport chain with shift to increase in glycolytic pathway. CONCLUSION VD supplementation led to cardiac remodeling and this process may be modulated by TXNIP and Trx proteins and consequently oxidative stress.
Collapse
Affiliation(s)
- Priscila P. dos Santos
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
- UNESPInstituto de Biociências de BotucatuBotucatuSPBrasilInstituto de Biociências de Botucatu-UNESP, Botucatu, SP - Brasil.
| | - Bruna P. M. Rafacho
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
| | - Andrea F. Gonçalves
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
| | - Vanessa C. M. Pires
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
| | - Meliza G. Roscani
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
| | - Paula S. Azevedo
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
| | - Bertha F. Polegato
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
| | - Marcos F. Minicucci
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
| | - Ana Angélica H. Fernandes
- UNESPInstituto de Biociências de BotucatuBotucatuSPBrasilInstituto de Biociências de Botucatu-UNESP, Botucatu, SP - Brasil.
| | - Suzana E. Tanni
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
| | - Leonardo A. M. Zornoff
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
| | - Sergio A. R. de Paiva
- UNESPFaculdade de Medicina de BotucatuBotucatuSPBrasilFaculdade de Medicina de Botucatu – UNESP, Botucatu, SP - Brasil.
- Centro de Pesquisa em AlimentosSão PauloSPBrasilCentro de Pesquisa em Alimentos, São Paulo, SP - Brasil.
| |
Collapse
|
17
|
Altun S, Budak H. The protective effect of the cardiac thioredoxin system on the heart in the case of iron overload in mice. J Trace Elem Med Biol 2021; 64:126704. [PMID: 33370714 DOI: 10.1016/j.jtemb.2020.126704] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 10/27/2020] [Accepted: 12/10/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND Iron, which is essential for many vital biological processes, causes significant clinical pathologies in the case of its deficiency or excess. Cardiovascular protective pathways are activated by iron therapy. However, determining the appropriate iron concentration is essential to protect heart tissue from iron-induced oxidative stress. The thioredoxin system is one of the antioxidant systems that protect cells against oxidative stress. Moreover, it allows the binding of many transcription factors for apoptosis, myocardial protection, the stimulation of cell proliferation, and angiogenesis processes, especially the regulation of the cardiovascular system. This study's goal was to understand how iron overload affects the gene and protein levels of the thioredoxin system in the mouse heart. METHODS BALB/c mice were randomly separated into two groups. The iron overload group was administered with intraperitoneal injections of an iron-dextran solution twice a week for three weeks. In parallel, the control group was intraperitoneally given Dextran 5 solution. The total iron content, the total GSH level, the reduced glutathione/oxidized glutathione (GSH/GSSG) ratio, and thioredoxin reductase 1 (TXNRD1) activity were demonstrated spectroscopically. Changes in the iron metabolism marker genes and thioredoxin system genes were examined by qPCR. The quantitative protein expression of TXNRD1 and thioredoxin-interacting protein (TXNIP) was examined by western blotting. RESULTS The iron content of the heart increased in the iron overload group. The expression of hepcidin (Hamp) and ferroportin (Fpn) increased with iron overload. However, decreased expression was observed for ferritin (Fth). No changes were revealed in the GSH level and GSH/GSSG ratio. The gene expression of thioredoxin 1 (Txn1), Txnrd1, and Txnip did not change. TXNRD1 activity and protein expression increased significantly, while the protein expression of TXNIP decreased significantly. CONCLUSION In the case of iron overload, the cardiac thioredoxin system is affected by the protein level rather than the gene level. The amount and duration of iron overload used in this study may be considered as a starting point for further studies to determine appropriate conditions for the iron therapy of cardiovascular diseases.
Collapse
Affiliation(s)
- Sevda Altun
- Science Faculty, Department of Molecular Biology and Genetics, Atatürk University, Erzurum, Turkey; Rafet Kayış Faculty of Engineering, Department of Genetic and Bioengineering, Alaaddin Keykubat University, Antalya, Turkey
| | - Harun Budak
- Science Faculty, Department of Molecular Biology and Genetics, Atatürk University, Erzurum, Turkey.
| |
Collapse
|
18
|
Nakayama Y, Mukai N, Wang BF, Yang K, Patwari P, Kitsis RN, Yoshioka J. Txnip C247S mutation protects the heart against acute myocardial infarction. J Mol Cell Cardiol 2021; 155:36-49. [PMID: 33652022 DOI: 10.1016/j.yjmcc.2021.02.013] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/11/2021] [Accepted: 02/20/2021] [Indexed: 01/15/2023]
Abstract
RATIONALE Thioredoxin-interacting protein (Txnip) is a novel molecular target with translational potential in diverse human diseases. Txnip has several established cellular actions including binding to thioredoxin, a scavenger of reactive oxygen species (ROS). It has been long recognized from in vitro evidence that Txnip forms a disulfide bridge through cysteine 247 (C247) with reduced thioredoxin to inhibit the anti-oxidative properties of thioredoxin. However, the physiological significance of the Txnip-thioredoxin interaction remains largely undefined in vivo. OBJECTIVE A single mutation of Txnip, C247S, abolishes the binding of Txnip with thioredoxin. Using a conditional and inducible approach with a mouse model of a mutant Txnip that does not bind thioredoxin, we tested whether the interaction of thioredoxin with Txnip is required for Txnip's pro-oxidative or cytotoxic effects in the heart. METHODS AND RESULTS Overexpression of Txnip C247S in cells resulted in a reduction in ROS, due to an inability to inhibit thioredoxin. Hypoxia (1% O2, 24 h)-induced killing effects of Txnip were decreased by lower levels of cellular ROS in Txnip C247S-expressing cells compared with wild-type Txnip-expressing cells. Then, myocardial ischemic injuries were assessed in the animal model. Cardiomyocyte-specific Txnip C247S knock-in mice had better survival with smaller infarct size following myocardial infarction (MI) compared to control animals. The absence of Txnip's inhibition of thioredoxin promoted mitochondrial anti-oxidative capacities in cardiomyocytes, thereby protecting the heart from oxidative damage induced by MI. Furthermore, an unbiased RNA sequencing screen identified that hypoxia-inducible factor 1 signaling pathway was involved in Txnip C247S-mediated cardioprotective mechanisms. CONCLUSION Txnip is a cysteine-containing redox protein that robustly regulates the thioredoxin system via a disulfide bond-switching mechanism in adult cardiomyocytes. Our results provide the direct in vivo evidence that regulation of redox state by Txnip is a crucial component for myocardial homeostasis under ischemic stress.
Collapse
Affiliation(s)
- Yoshinobu Nakayama
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, NY, New York, United States of America
| | - Nobuhiro Mukai
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, NY, New York, United States of America
| | - Bing F Wang
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Kristen Yang
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, NY, New York, United States of America
| | - Parth Patwari
- Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States of America
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, Bronx, NY, United States of America
| | - Jun Yoshioka
- Department of Molecular, Cellular & Biomedical Sciences, City University of New York School of Medicine, City College of New York, NY, New York, United States of America; Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, United States of America.
| |
Collapse
|
19
|
Zavala MR, Díaz RG, Villa-Abrille MC, Pérez NG. Thioredoxin 1 (TRX1) Overexpression Cancels the Slow Force Response (SFR) Development. Front Cardiovasc Med 2021; 8:622583. [PMID: 33718450 PMCID: PMC7952880 DOI: 10.3389/fcvm.2021.622583] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 01/18/2021] [Indexed: 11/28/2022] Open
Abstract
The stretch of cardiac muscle increases developed force in two phases. The first phase occurs immediately after stretch and is the expression of the Frank–Starling mechanism, while the second one or slow force response (SFR) occurs gradually and is due to an increase in the calcium transient amplitude. An important step in the chain of events leading to the SFR generation is the increased production of reactive oxygen species (ROS) leading to redox sensitive ERK1/2, p90RSK, and NHE1 phosphorylation/activation. Conversely, suppression of ROS production blunts the SFR. The purpose of this study was to explore whether overexpression of the ubiquitously expressed antioxidant molecule thioredoxin-1 (TRX1) affects the SFR development and NHE1 phosphorylation. We did not detect any change in basal phopho-ERK1/2, phopho-p90RSK, and NHE1 expression in mice with TRX1 overexpression compared to wild type (WT). Isolated papillary muscles from WT or TRX1-overexpressing mice were stretched from 92 to 98% of its maximal length. A prominent SFR was observed in WT mice that was completely canceled in TRX1 animals. Interestingly, myocardial stretch induced a significant increase in NHE1 phosphorylation in WT mice that was not detected in TRX1-overexpressing mice. These novel results suggest that magnification of cardiac antioxidant defense power by overexpression of TRX1 precludes NHE1 phosphorylation/activation after stretch, consequently blunting the SFR development.
Collapse
Affiliation(s)
- Maite R Zavala
- Fellow From Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Buenos Aires, Argentina
| | - Romina G Díaz
- Established Investigators of CONICET, Buenos Aires, Argentina
| | - María C Villa-Abrille
- Established Investigators of CONICET, Buenos Aires, Argentina.,Full Professors of Physiology, Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, Buenos Aires, Argentina
| | - Néstor G Pérez
- Established Investigators of CONICET, Buenos Aires, Argentina.,Full Professors of Physiology, Facultad de Ciencias Médicas de La Plata, Universidad Nacional de La Plata, Buenos Aires, Argentina
| |
Collapse
|
20
|
Cortassa S, Juhaszova M, Aon MA, Zorov DB, Sollott SJ. Mitochondrial Ca 2+, redox environment and ROS emission in heart failure: Two sides of the same coin? J Mol Cell Cardiol 2021; 151:113-125. [PMID: 33301801 PMCID: PMC7880885 DOI: 10.1016/j.yjmcc.2020.11.013] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 11/05/2020] [Accepted: 11/28/2020] [Indexed: 12/11/2022]
Abstract
Heart failure (HF) is a progressive, debilitating condition characterized, in part, by altered ionic equilibria, increased ROS production and impaired cellular energy metabolism, contributing to variable profiles of systolic and diastolic dysfunction with significant functional limitations and risk of premature death. We summarize current knowledge concerning changes of intracellular Na+ and Ca2+ control mechanisms during the disease progression and their consequences on mitochondrial Ca2+ homeostasis and the shift in redox balance. Absent existing biological data, our computational modeling studies advance a new 'in silico' analysis to reconcile existing opposing views, based on different experimental HF models, regarding variations in mitochondrial Ca2+ concentration that participate in triggering and perpetuating oxidative stress in the failing heart and their impact on cardiac energetics. In agreement with our hypothesis and the literature, model simulations demonstrate the possibility that the heart's redox status together with cytoplasmic Na+ concentrations act as regulators of mitochondrial Ca2+ levels in HF and of the bioenergetics response that will ultimately drive ATP supply and oxidative stress. The resulting model predictions propose future directions to study the evolution of HF as well as other types of heart disease, and to develop novel testable mechanistic hypotheses that may lead to improved therapeutics.
Collapse
Affiliation(s)
- Sonia Cortassa
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States.
| | - Magdalena Juhaszova
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States.
| | - Miguel A Aon
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States; Translational Gerontology Branch, National Institute on Aging, NIH, Baltimore, MD, United States.
| | - Dmitry B Zorov
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia.
| | - Steven J Sollott
- Laboratory of Cardiovascular Science, National Institute on Aging, NIH, Baltimore, MD, United States.
| |
Collapse
|
21
|
Ralston NV, Raymond LJ. Soft electrophile inhibition of selenoenzymes in disease pathologies. Toxicology 2021. [DOI: 10.1016/b978-0-12-819092-0.00040-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
22
|
Dunn S, Hilgers RH, Das KC. Thioredoxin deficiency exacerbates vascular dysfunction during diet-induced obesity in small mesenteric artery in mice. Microcirculation 2020; 28:e12674. [PMID: 33316843 DOI: 10.1111/micc.12674] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Accepted: 12/07/2020] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Thioredoxin (Trx) is a small cellular redox protein with established antioxidant and disulfide reductase properties. We hypothesized that Trx deficiency in mice would cause increased oxidative stress with consequent redox imbalance that would exacerbate obesity-induced vascular dysfunction. METHODS Non-transgenic (NT, C57BL/6) and dominant-negative Trx (dnTrx-Tg, low levels of redox-active protein) mice were either fed a normal diet (NC) or high fat diet plus sucrose (HFS) diet for 4 months (3-month HFD+ 1-month HFS). Weight gain, glucose tolerance test (GTT), insulin tolerance test (ITT), and other metabolic parameters were performed following NC or HFS diet. Arterial structural remodeling and functional parameters were assessed by myography. RESULTS Our study found that dnTrx mice with lower levels of active Trx exacerbated myogenic tone, inward arterial remodeling, arterial stiffening, phenylephrine-induced contraction, and endothelial dysfunction of MA. Additionally, FeTMPyP, a peroxynitrite decomposition catalyst, acutely decreased myogenic tone and contraction and normalized endothelial function in MA from dnTrx-Tg mice on HFS via increasing nitric oxide (NO)-mediated relaxation. CONCLUSIONS Our results indicate that deficiency of active Trx exacerbates MA contractile and relaxing properties during diet-induced obesity demonstrating that loss of redox balance in obesity is a key mechanism of vascular endothelial dysfunction.
Collapse
Affiliation(s)
- Shannon Dunn
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Robert H Hilgers
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - Kumuda C Das
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
23
|
Oxidative Stress and Antioxidant Treatments in Cardiovascular Diseases. Antioxidants (Basel) 2020; 9:antiox9121292. [PMID: 33348578 PMCID: PMC7766219 DOI: 10.3390/antiox9121292] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 12/04/2020] [Accepted: 12/10/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress plays a key role in many physiological and pathological conditions. The intracellular oxidative homeostasis is tightly regulated by the reactive oxygen species production and the intracellular defense mechanisms. Increased oxidative stress could alter lipid, DNA, and protein, resulting in cellular inflammation and programmed cell death. Evidences show that oxidative stress plays an important role in the progression of various cardiovascular diseases, such as atherosclerosis, heart failure, cardiac arrhythmia, and ischemia-reperfusion injury. There are a number of therapeutic options to treat oxidative stress-associated cardiovascular diseases. Well known antioxidants, such as nutritional supplements, as well as more novel antioxidants have been studied. In addition, novel therapeutic strategies using miRNA and nanomedicine are also being developed to treat various cardiovascular diseases. In this article, we provide a detailed description of oxidative stress. Then, we will introduce the relationship between oxidative stress and several cardiovascular diseases. Finally, we will focus on the clinical implications of oxidative stress in cardiovascular diseases.
Collapse
|
24
|
Demarchi A, Somaschini A, Cornara S, Androulakis E. Peripheral Artery Disease in Diabetes Mellitus: Focus on Novel Treatment Options. Curr Pharm Des 2020; 26:5953-5968. [PMID: 33243109 DOI: 10.2174/1389201021666201126143217] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 08/09/2020] [Indexed: 02/07/2023]
Abstract
Diabetes mellitus (DM) and peripheral artery disease (PAD) are two clinical entities closely associated. They share many pathophysiological pathways such as inflammation, endothelial dysfunction, oxidative stress and pro-coagulative unbalance. Emerging data focusing on agents targeting these pathways may be promising. Moreover, due to the increased cardiovascular risk, there is a growing interest in cardiovascular and "pleiotropic" effects of novel glucose lowering drugs. This review summarizes the main clinical features of PAD in patients, the diagnostic process and current medical/interventional approaches, ranging from "classical treatment" to novel agents.
Collapse
Affiliation(s)
| | - Alberto Somaschini
- Adult Intensive Care Unit, Royal Brompton & Harefield NHS Foundation Trust, London, United Kingdom
| | | | - Emmanuel Androulakis
- Adult Intensive Care Unit, Royal Brompton & Harefield NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
25
|
Cai M, Xu Z, Bo W, Wu F, Qi W, Tian Z. Up-regulation of Thioredoxin 1 by aerobic exercise training attenuates endoplasmic reticulum stress and cardiomyocyte apoptosis following myocardial infarction. SPORTS MEDICINE AND HEALTH SCIENCE 2020; 2:132-140. [PMID: 35782283 PMCID: PMC9219273 DOI: 10.1016/j.smhs.2020.07.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 11/13/2022] Open
Abstract
Exercise training (ET) has been reported to reduce oxidative stress and endoplasmic reticulum (ER) stress in the heart following myocardial infarction (MI). Thioredoxin 1 (Trx1) plays a protective role in the infarcted heart. However, whether Trx1 regulates ER stress of the infarcted heart and participates in ET-induced cardiac protective effects are still not well known. In this work, H9c2 cells were treated with hydrogen peroxide (H2O2) and recombinant human Trx1 protein (TXN), meanwhile, adult male C57B6L mice were used to establish the MI model, and subjected to a six-week aerobic exercise training (AET) with or without the injection of Trx1 inhibitor, PX-12. Results showed that H2O2 significantly increased reactive oxygen species (ROS) level and the expression of TXNIP, CHOP and cleaved caspase12, induced cell apoptosis; TXN intervention reduced ROS level and the expression of CHOP and cleaved caspase12, and inhibited cell apoptosis in H2O2-treated H9c2 cells. Furthermore, AET up-regulated endogenous Trx1 protein expression and down-regulated TXNIP expression, restored ROS level and the expression of ER stress-related proteins, inhibited cell apoptosis as well as improved cardiac fibrosis and heart function in mice after MI. PX-12 partly inhibited the AET-induced beneficial effects in the infarcted heart. This study demonstrates that Trx1 attenuates ER stress-induced cell apoptosis, and AET reduces MI-induced ROS overproduction, ER stress and cell apoptosis partly through up-regulating of Trx1 expression in mice with MI.
Collapse
|
26
|
Affiliation(s)
- Francesco Perone
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Via dell'Elettronica, 86077 Pozzilli (IS), Italy
| | - Giuseppe Lembo
- Department of Angiocardioneurology and Translational Medicine, IRCCS Neuromed, Via dell'Elettronica, 86077 Pozzilli (IS), Italy.,Department of Molecular Medicine, "Sapienza" University of Rome, Rome, Italy
| |
Collapse
|
27
|
Khan S, Ahmad SS, Kamal MA. Diabetic Cardiomyopathy: From Mechanism to Management in a Nutshell. Endocr Metab Immune Disord Drug Targets 2020; 21:268-281. [PMID: 32735531 DOI: 10.2174/1871530320666200731174724] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/23/2019] [Revised: 06/03/2020] [Accepted: 07/06/2020] [Indexed: 11/22/2022]
Abstract
Diabetic cardiomyopathy (DCM) is a significant complication of diabetes mellitus characterized by gradually failing heart with detrimental cardiac remodelings, such as fibrosis and diastolic and systolic dysfunction, which is not directly attributable to coronary artery disease. Insulin resistance and resulting hyperglycemia is the main trigger involved in the initiation of diabetic cardiomyopathy. There is a constellation of many pathophysiological events, such as lipotoxicity, oxidative stress, inflammation, inappropriate activation of the renin-angiotensin-aldosterone system, dysfunctional immune modulation promoting increased rate of cardiac cell injury, apoptosis, and necrosis, which ultimately culminates into interstitial fibrosis, cardiac stiffness, diastolic dysfunction, initially, and later systolic dysfunction too. These events finally lead to clinical heart failure of DCM. Herein, The pathophysiology of DCM is briefly discussed. Furthermore, potential therapeutic strategies currently used for DCM are also briefly mentioned.
Collapse
Affiliation(s)
- Shahzad Khan
- Department of Pathophysiology, Wuhan University School of Medicine, Hubei, Wuhan, China
| | - Syed S Ahmad
- Department of Bioengineering, Faculty of Engineering, Integral University, Lucknow, India
| | - Mohammad A Kamal
- King Fahd Medical Research Center, King Abdulaziz University, P.O. Box 80216, Jeddah 21589, Saudi Arabia
| |
Collapse
|
28
|
Yagisawa Y, Suita K, Ohnuki Y, Ishikawa M, Mototani Y, Ito A, Matsuo I, Hayakawa Y, Nariyama M, Umeki D, Saeki Y, Amitani Y, Nakamura Y, Tomonari H, Okumura S. Effects of occlusal disharmony on cardiac fibrosis, myocyte apoptosis and myocyte oxidative DNA damage in mice. PLoS One 2020; 15:e0236547. [PMID: 32716920 PMCID: PMC7384634 DOI: 10.1371/journal.pone.0236547] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 07/08/2020] [Indexed: 12/05/2022] Open
Abstract
Occlusal disharmony leads to morphological changes in the hippocampus and osteopenia of the lumbar vertebra and long bones in mice, and causes stress. Various types of stress are associated with increased incidence of cardiovascular disease, but the relationship between occlusal disharmony and cardiovascular disease remain poorly understood. Therefore, in this work, we examined the effects of occlusal disharmony on cardiac homeostasis in bite-opening (BO) mice, in which a 0.7 mm space was introduced by cementing a suitable applicance onto the mandibular incisior. We first examined the effects of BO on the level of serum corticosterone, a key biomarker for stress, and on heart rate variability at 14 days after BO treatment, compared with baseline. BO treatment increased serum corticosterone levels by approximately 3.6-fold and the low frequency/high frequency ratio, an index of sympathetic nervous activity, was significantly increased by approximately 4-fold by the BO treatment. We then examined the effects of BO treatment on cardiac homeostasis in mice treated or not treated with the non-selective β-blocker propranolol for 2 weeks. Cardiac function was significantly decreased in the BO group compared to the control group, but propranolol ameliorated the dysfunction. Cardiac fibrosis, myocyte apoptosis and myocyte oxidative DNA damage were significantly increased in the BO group, but propranolol blocked these changes. The BO-induced cardiac dysfunction was associated with increased phospholamban phosphorylation at threonine-17 and serine-16, as well as inhibition of Akt/mTOR signaling and autophagic flux. These data suggest that occlusal disharmony might affect cardiac homeostasis via alteration of the autonomic nervous system.
Collapse
Affiliation(s)
- Yuka Yagisawa
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Kenji Suita
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Ohnuki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Misao Ishikawa
- Department of Oral Anatomy, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasumasa Mototani
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Aiko Ito
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Ichiro Matsuo
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Periodontology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshio Hayakawa
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
- Department of Dental Anesthesiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Megumi Nariyama
- Department of Pediatric Dentistry, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Daisuke Umeki
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasutake Saeki
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yasuharu Amitani
- Department of Mathematics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Yoshiki Nakamura
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Hiroshi Tomonari
- Department of Orthodontics, Tsurumi University School of Dental Medicine, Yokohama, Japan
| | - Satoshi Okumura
- Department of Physiology, Tsurumi University School of Dental Medicine, Yokohama, Japan
| |
Collapse
|
29
|
Liu F, Su H, Liu B, Mei Y, Ke Q, Sun X, Tan W. STVNa Attenuates Isoproterenol-Induced Cardiac Hypertrophy Response through the HDAC4 and Prdx2/ROS/Trx1 Pathways. Int J Mol Sci 2020; 21:ijms21020682. [PMID: 31968660 PMCID: PMC7014432 DOI: 10.3390/ijms21020682] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 01/10/2020] [Accepted: 01/11/2020] [Indexed: 12/13/2022] Open
Abstract
Recent data show that cardiac hypertrophy contributes substantially to the overall heart failure burden. Mitochondrial dysfunction is a common feature of cardiac hypertrophy. Recent studies have reported that isosteviol inhibits myocardial ischemia-reperfusion injury in guinea pigs and H9c2 cells. This work investigated the protective mechanisms of isosteviol sodium (STVNa) against isoproterenol (Iso)-induced cardiac hypertrophy. We found that STVNa significantly inhibited H9c2 cell and rat primary cardiomyocyte cell surface, restored mitochondrial membrane potential (MMP) and morphological integrity, and decreased the expression of mitochondrial function-related proteins Fis1 and Drp1. Furthermore, STVNa decreased reactive oxygen species (ROS) levels and upregulated the expression of antioxidant factors, Thioredoxin 1 (Trx1) and Peroxiredoxin 2 (Prdx2). Moreover, STVNa restored the activity of histone deacetylase 4 (HDAC4) in the nucleus. Together, our data show that STVNa confers protection against Iso-induced myocardial hypertrophy primarily through the Prdx2/ROS/Trx1 signaling pathway. Thus, STVNA is a potentially effective treatment for cardiac hypertrophy in humans.
Collapse
Affiliation(s)
| | | | | | | | | | - Xiaoou Sun
- Correspondence: (X.S.); (W.T.); Tel.: +86-13539850005 (X.S.); +86-13928954505 (W.T.)
| | - Wen Tan
- Correspondence: (X.S.); (W.T.); Tel.: +86-13539850005 (X.S.); +86-13928954505 (W.T.)
| |
Collapse
|
30
|
Hemling P, Zibrova D, Strutz J, Sohrabi Y, Desoye G, Schulten H, Findeisen H, Heller R, Godfrey R, Waltenberger J. Hyperglycemia-induced endothelial dysfunction is alleviated by thioredoxin mimetic peptides through the restoration of VEGFR-2-induced responses and improved cell survival. Int J Cardiol 2019; 308:73-81. [PMID: 31955977 DOI: 10.1016/j.ijcard.2019.12.065] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 12/05/2019] [Accepted: 12/29/2019] [Indexed: 01/16/2023]
Abstract
BACKGROUND Diabetes mellitus is an important cardiovascular risk factor characterized by elevated plasma glucose levels. High glucose (HG) negatively influences endothelial cell (EC) function, which is characterized by the inability of ECs to respond to vascular endothelial growth factor (VEGF-A) stimulation. We aimed to identify potential strategies to improve EC function in diabetes. METHODS AND RESULTS Human umbilical cord endothelial cells (HUVECs) were subjected to hyperglycemic milieu by exposing cells to HG together with glucose metabolite, methylglyoxal (MG) in vitro. Hyperglycemic cells showed reduced chemotactic responses towards VEGF-A as revealed by Boyden chamber migration assays, indicating the development of "VEGF resistance" phenotype. Furthermore, HG/MG-exposed cells were defective in their general migratory and proliferative responses and were in a pro-apoptotic state. Mechanistically, the exposure to HG/MG resulted in reactive oxygen species (ROS) accumulation which is secondary to the impairment of thioredoxin (Trx) activity in these cells. Pharmacological and genetic targeting of Trx recapitulated VEGF resistance. Functional supplementation of Trx using thioredoxin mimetic peptides (TMP) reversed the HG/MG-induced ROS generation, improved the migration, proliferation, survival and restored VEGF-A-induced chemotaxis and sprouting angiogenesis of hyperglycemic ECs. Importantly, TMP treatment reduced ROS accumulation and improved VEGF-A responses of placental arterial endothelial cells isolated from gestational diabetes mellitus patients. CONCLUSIONS Our findings suggest a putative role for Trx in modulating EC function and its functional impairment in HG conditions contribute to EC dysfunction. Supplementation of TMP could be used as a novel strategy to improve endothelial cell function in diabetes.
Collapse
Affiliation(s)
- Pia Hemling
- Experimental and Molecular Cardiology, Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany; Molecular Cardiology, Department of Cardiology I - Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany; Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Münster, Germany
| | - Darya Zibrova
- Institute for Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital Jena, Jena, Germany
| | - Jasmin Strutz
- Department of Obstetrics and Gynecology, Medical University of Graz, Austria
| | - Yahya Sohrabi
- Molecular Cardiology, Department of Cardiology I - Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| | - Gernot Desoye
- Department of Obstetrics and Gynecology, Medical University of Graz, Austria
| | - Henny Schulten
- Experimental and Molecular Cardiology, Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany; Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, the Netherlands
| | - Hannes Findeisen
- Molecular Cardiology, Department of Cardiology I - Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany
| | - Regine Heller
- Institute for Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital Jena, Jena, Germany
| | - Rinesh Godfrey
- Experimental and Molecular Cardiology, Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany; Molecular Cardiology, Department of Cardiology I - Coronary and Peripheral Vascular Disease, Heart Failure, University Hospital Münster, Münster, Germany; Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, the Netherlands; Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Münster, Germany.
| | - Johannes Waltenberger
- Experimental and Molecular Cardiology, Department of Cardiovascular Medicine, University Hospital Münster, Münster, Germany; Department of Physiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht, the Netherlands; Cells-in-Motion Cluster of Excellence (EXC 1003-CiM), University of Münster, Münster, Germany; Department of Internal Medicine I, SRH Central Hospial, Suhl, Germany.
| |
Collapse
|
31
|
Zhou R, Gao J, Xiang C, Liu Z, Zhang Y, Zhang J, Yang H. Salvianolic acid A attenuated myocardial infarction–induced apoptosis and inflammation by activating Trx. Naunyn Schmiedebergs Arch Pharmacol 2019; 393:991-1002. [DOI: 10.1007/s00210-019-01766-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Accepted: 11/08/2019] [Indexed: 01/30/2023]
|
32
|
Rocca C, Pasqua T, Boukhzar L, Anouar Y, Angelone T. Progress in the emerging role of selenoproteins in cardiovascular disease: focus on endoplasmic reticulum-resident selenoproteins. Cell Mol Life Sci 2019; 76:3969-3985. [PMID: 31218451 PMCID: PMC11105271 DOI: 10.1007/s00018-019-03195-1] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 05/29/2019] [Accepted: 06/14/2019] [Indexed: 12/30/2022]
Abstract
Cardiovascular diseases represent one of the most important health problems of developed countries. One of the main actors involved in the onset and development of cardiovascular diseases is the increased production of reactive oxygen species that, through lipid peroxidation, protein oxidation and DNA damage, induce oxidative stress and cell death. Basic and clinical research are ongoing to better understand the endogenous antioxidant mechanisms that counteract oxidative stress, which may allow to identify a possible therapeutic targeting/application in the field of stress-dependent cardiovascular pathologies. In this context, increasing attention is paid to the glutathione/glutathione-peroxidase and to the thioredoxin/thioredoxin-reductase systems, among the most potent endogenous antioxidative systems. These key enzymes, belonging to the selenoprotein family, have a well-established function in the regulation of the oxidative cell balance. The aim of the present review was to highlight the role of selenoproteins in cardiovascular diseases, introducing the emerging cardioprotective role of endoplasmic reticulum-resident members and in particular one of them, namely selenoprotein T or SELENOT. Accumulating evidence indicates that the dysfunction of different selenoproteins is involved in the susceptibility to oxidative stress and its associated cardiovascular alterations, such as congestive heart failure, coronary diseases, impaired cardiac structure and function. Some of them are under investigation as useful pathological biomarkers. In addition, SELENOT exhibited intriguing cardioprotective effects by reducing the cardiac ischemic damage, in terms of infarct size and performance. In conclusion, selenoproteins could represent valuable targets to treat and diagnose cardiovascular diseases secondary to oxidative stress, opening a new avenue in the field of related therapeutic strategies.
Collapse
Affiliation(s)
- Carmine Rocca
- Laboratory of Cellular and Molecular Cardiovascular Patho-physiology, Department of Biology, E. and E.S., University of Calabria, Rende, Italy.
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen-Normandie University, 76821, Mont-Saint-Aignan, France.
- Institute for Research and Innovation in Biomedicine, 76000, Rouen, France.
| | - Teresa Pasqua
- Laboratory of Cellular and Molecular Cardiovascular Patho-physiology, Department of Biology, E. and E.S., University of Calabria, Rende, Italy
- "Fondazione Umberto Veronesi", Milan, Italy
| | - Loubna Boukhzar
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen-Normandie University, 76821, Mont-Saint-Aignan, France
- Institute for Research and Innovation in Biomedicine, 76000, Rouen, France
| | - Youssef Anouar
- UNIROUEN, Inserm U1239, Neuronal and Neuroendocrine Differentiation and Communication Laboratory, Rouen-Normandie University, 76821, Mont-Saint-Aignan, France
- Institute for Research and Innovation in Biomedicine, 76000, Rouen, France
| | - Tommaso Angelone
- Laboratory of Cellular and Molecular Cardiovascular Patho-physiology, Department of Biology, E. and E.S., University of Calabria, Rende, Italy.
- National Institute of Cardiovascular Research (INRC), Bologna, Italy.
| |
Collapse
|
33
|
Marschner RA, Banda P, Wajner SM, Markoski MM, Schaun M, Lehnen AM. Short-term exercise training improves cardiac function associated to a better antioxidant response and lower type 3 iodothyronine deiodinase activity after myocardial infarction. PLoS One 2019; 14:e0222334. [PMID: 31513640 PMCID: PMC6742396 DOI: 10.1371/journal.pone.0222334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/27/2019] [Indexed: 12/11/2022] Open
Abstract
Aims We assessed the effects of a short-term exercise training on cardiac function, oxidative stress markers, and type 3 iodothyronine deiodinase (D3) activity in cardiac tissue of spontaneously hypertensive rats (SHR) following experimental myocardial infarction (MI). Methods Twenty-four SHR (aged 3 months) were allocated to 4 groups: sham+sedentary, sham+trained, MI+sedentary and MI+trained. MI was performed by permanent ligation of the coronary artery. Exercise training (treadmill) started 96 hours after MI and lasted for 4 weeks (~60% maximum effort, 4x/week and 40 min/day). Cardiac function (echocardiography), thioredoxin reductase (TRx), total carbonyl levels, among other oxidative stress markers and D3 activity were measured. A Generalized Estimating Equation was used, followed by Bonferroni’s test (p<0.05). Results MI resulted in an increase in left ventricular mass (p = 0.002) with decreased cardiac output (~22.0%, p = 0.047) and decreased ejection fraction (~41%, p = 0.008) as well as an increase in the carbonyl levels (p = 0.001) and D3 activity (~33%, p<0.001). Exercise training resulted in a decrease in left ventricular mass, restored cardiac output (~34%, p = 0.048) and ejection fraction (~20%, p = 0.040), increased TRx (~85%, p = 0.007) and reduced carbonyl levels (p<0.001) and D3 activity (p<0.001). Conclusions Our short-term exercise training helped reverse the effects of MI on cardiac function. These benefits seem to derive from a more efficient antioxidant response and lower D3 activity in cardiac tissue.
Collapse
Affiliation(s)
- Rafael Aguiar Marschner
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology, Porto Alegre, Rio Grande do Sul, Brazil
- Thyroid Division, Endocrinology Service, Hospital de Clínicas de Porto Alegre/Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Patrícia Banda
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Simone Magagnin Wajner
- Thyroid Division, Endocrinology Service, Hospital de Clínicas de Porto Alegre/Federal University of Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | | | - Maximiliano Schaun
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology, Porto Alegre, Rio Grande do Sul, Brazil
| | - Alexandre Machado Lehnen
- Institute of Cardiology of Rio Grande do Sul/University Foundation of Cardiology, Porto Alegre, Rio Grande do Sul, Brazil
- * E-mail:
| |
Collapse
|
34
|
Zaobornyj T, Mazo T, Perez V, Gomez A, Contin M, Tripodi V, D'Annunzio V, Gelpi RJ. Thioredoxin-1 is required for the cardioprotecive effect of sildenafil against ischaemia/reperfusion injury and mitochondrial dysfunction in mice. Free Radic Res 2019; 53:993-1004. [PMID: 31455116 DOI: 10.1080/10715762.2019.1661404] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Sildenafil is a phosphodiesterase type 5 inhibitor which confers cardioprotection against myocardial ischaemia/reperfusion (I/R) injury. The aim of this study was to determine if Trx1 participates in cardioprotection exerted by sildenafil in an acute model of I/R, and to evaluate mitochondrial bioenergetics and cellular redox status. Langendorff-perfused hearts from wild type (WT) mice and a dominant negative (DN-Trx1) mutant of Trx1 were assigned to placebo or sildenafil (0.7 mg/kg i.p.) and subjected to 30 min of ischaemia followed by 120 min of reperfusion. WT + S showed a significant reduction of infarct size (51.2 ± 3.0% vs. 30 ± 3.0%, p < .001), an effect not observed in DN-Trx. After I/R, sildenafil preserved state 3 oxygen consumption from WT, but had a milder effect in DN-Trx1 only partially protecting state 3 values. Treatment restored respiratory control (RC) after I/R, which resulted 8% (WT) and 24% (DN-Trx1) lower than in basal conditions. After I/R, a significant increase in H2O2 production was observed both for WT and DN-Trx (WT: 1.17 ± 0.13 nmol/mg protein and DN-Trx: 1.38 ± 0.12 nmol/min mg protein). With sildenafil, values were 21% lower only in WT I/R. Treatment decreased GSSG levels both in WT and DN-Trx1. In addition, GSSG/GSH2 ratio was partially restored by sildenafil. Also, an increase in p-eNOS/eNOS even before the myocardial ischaemia was observed with sildenafil, both in WT (14%, p > .05) and in DN-Trx (35%, p < .05). Active Trx1 is required for the onset of the cardioprotective effects of sildenafil on I/R injury, together with the preservation of cellular redox balance and mitochondrial function.
Collapse
Affiliation(s)
- Tamara Zaobornyj
- Department of Analytical Chemistry and Physical Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires , Buenos Aires , Argentina.,Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET) , Buenos Aires , Argentina.,National Council of Scientific and Technical Research (CONICET) , Buenos Aires , Argentina
| | - Tamara Mazo
- Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET) , Buenos Aires , Argentina
| | - Virginia Perez
- Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET) , Buenos Aires , Argentina.,Department of Pathology, Faculty of Medicine, Institute of Cardiovascular Physiopathology, University of Buenos Aires , Buenos Aires , Argentina
| | - Anabella Gomez
- Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET) , Buenos Aires , Argentina.,Department of Pathology, Faculty of Medicine, Institute of Cardiovascular Physiopathology, University of Buenos Aires , Buenos Aires , Argentina
| | - Mario Contin
- Department of Analytical Chemistry and Physical Chemistry, School of Pharmacy and Biochemistry, University of Buenos Aires , Buenos Aires , Argentina
| | - Valeria Tripodi
- National Council of Scientific and Technical Research (CONICET) , Buenos Aires , Argentina.,Department of Pharmaceutical Technology, School of Pharmacy and Biochemistry, University of Buenos Aires , Buenos Aires , Argentina
| | - Verónica D'Annunzio
- Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET) , Buenos Aires , Argentina.,National Council of Scientific and Technical Research (CONICET) , Buenos Aires , Argentina.,Department of Pathology, Faculty of Medicine, Institute of Cardiovascular Physiopathology, University of Buenos Aires , Buenos Aires , Argentina
| | - Ricardo J Gelpi
- Institute of Biochemistry and Molecular Medicine (IBIMOL UBA-CONICET) , Buenos Aires , Argentina.,National Council of Scientific and Technical Research (CONICET) , Buenos Aires , Argentina.,Department of Pathology, Faculty of Medicine, Institute of Cardiovascular Physiopathology, University of Buenos Aires , Buenos Aires , Argentina
| |
Collapse
|
35
|
Acute fluoride exposure alters myocardial redox and inflammatory markers in rats. Mol Biol Rep 2019; 46:6155-6164. [DOI: 10.1007/s11033-019-05050-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/28/2019] [Indexed: 01/31/2023]
|
36
|
Hofmanis J, Hofmane D, Svirskis S, Mackevics V, Tretjakovs P, Lejnieks A, Signorelli SS. HDL-C Role in Acquired Aortic Valve Stenosis Patients and Its Relationship with Oxidative Stress. ACTA ACUST UNITED AC 2019; 55:medicina55080416. [PMID: 31362438 PMCID: PMC6723197 DOI: 10.3390/medicina55080416] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/23/2019] [Accepted: 07/25/2019] [Indexed: 12/13/2022]
Abstract
Background and objectives: Mechanical stress is currently considered as the main factor promoting calcific aortic valve stenosis (AS) onset. It causes endothelial damage and dysfunction. The chronic inflammatory process causes oxidative stress. Oxidative stress-induced high-density lipoprotein cholesterol (HDL-C) dysfunction is an important component of the development of AS. The aim of the study was to evaluate the role of HDL-C in AS patients in three severity grades and in relation to the biomarkers of oxidative stress, thioredoxin reductase 1 (TrxR1) and myeloperoxidase (MPO). Materials and Methods: 18 patients with mild, 19 with moderate. and 15 with severe AS were included in the study, and 50 individuals were enrolled in the control group. Stenosis severity was determined by echocardiography. The TrxR1 and MPO were analyzed by ELISA, and HDL-C by commercially available tests. Data were analyzed using GraphPad Prism 8. Results: HDL-C in AS patients vs. control substantially decreases and this decline was observed in all three AS severity groups: mild (p = 0.018), moderate (p = 0.0002), and severe (p = 0.004). In both the control and the stenosis group, the HDL-C was higher in women than in men. In comparison to control, the HDL-C level was lower in the AS group, and more pronounced in women (p = 0.0001) than in men (p = 0.049). A higher TrxR1 level was observed in patients with mild (p = 0.0001) and severe AS (p = 0.047). However, a clear correlation between TrxR1 and HDL-C was not obtained. Analysis of MPO showed differences in all severity grades vs. control (p = 0.024 mild stenosis; p = 0.002 moderate stenosis; p = 0.0015 severe stenosis). A negative correlation (p = 0.047; rp = -0.28) was found between MPO and HDL-C, which confirms the adverse effects of MPO resulting in HDL-C dysfunction. Conclusions: In this study, we justified HDL-C level association with AS development process. The results unequivocally substantiated the association between HDL-C and AS in all severity grades in women, but only in moderate AS for men, which we explained by the small number of men in the groups. The obtained correlation between the HDL-C and MPO levels, as well as the concurrent decrease in the HDL-C level and increase in the TrxR1 level, indicate in general an HDL-C association with oxidative stress in AS patients.
Collapse
Affiliation(s)
- Juris Hofmanis
- Faculty of Medicine, Department of Internal Diseases, Riga Stradins University and Riga East University Hospital, LV-1007 Riga, Latvia
- Correspondence: (J.H.); (V.M.); Tel.: +371-28446644 (J.H.); +371-27540908 (V.M.)
| | | | - Simons Svirskis
- Institute of Microbiology and Virology, Riga Stradins University, LV-1007 Riga, Latvia
| | - Vitolds Mackevics
- Faculty of Medicine, Department of Internal Diseases, Riga Stradins University and Riga East University Hospital, LV-1007 Riga, Latvia
- Correspondence: (J.H.); (V.M.); Tel.: +371-28446644 (J.H.); +371-27540908 (V.M.)
| | - Peteris Tretjakovs
- Faculty of Medicine, Department of Human Physiology and Biochemistry, Riga Stradins University, LV-1007 Riga, Latvia
| | - Aivars Lejnieks
- Faculty of Medicine, Department of Internal Diseases, Riga Stradins University and Riga East University Hospital, LV-1007 Riga, Latvia
| | - Salvatore Santo Signorelli
- Department of Clinical and Experimental Medicine, University of Catania and c/o University Hospital “G.Rodolico”, 95123 Catania, Italy
| |
Collapse
|
37
|
Raffaeli G, Ghirardello S, Passera S, Mosca F, Cavallaro G. Oxidative Stress and Neonatal Respiratory Extracorporeal Membrane Oxygenation. Front Physiol 2018; 9:1739. [PMID: 30564143 PMCID: PMC6288438 DOI: 10.3389/fphys.2018.01739] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 11/19/2018] [Indexed: 12/16/2022] Open
Abstract
Oxidative stress is a frequent condition in critically ill patients, especially if exposed to extracorporeal circulation, and it is associated with worse outcomes and increased mortality. The inflammation triggered by the contact of blood with a non-endogenous surface, the use of high volumes of packed red blood cells and platelets transfusion, the risk of hyperoxia and the impairment of antioxidation systems contribute to the increase of reactive oxygen species and the imbalance of the redox system. This is responsible for the increased production of superoxide anion, hydrogen peroxide, hydroxyl radicals, and peroxynitrite resulting in increased lipid peroxidation, protein oxidation, and DNA damage. The understanding of the pathophysiologic mechanisms leading to redox imbalance would pave the way for the future development of preventive approaches. This review provides an overview of the clinical impact of the oxidative stress during neonatal extracorporeal support and concludes with a brief perspective on the current antioxidant strategies, with the aim to focus on the potential oxidative stress-mediated cell damage that has been implicated in both short and long-term outcomes.
Collapse
Affiliation(s)
- Genny Raffaeli
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Stefano Ghirardello
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Sofia Passera
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Fabio Mosca
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Giacomo Cavallaro
- NICU, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
38
|
Majumder A, Singh M, George AK, Tyagi SC. Restoration of skeletal muscle homeostasis by hydrogen sulfide during hyperhomocysteinemia-mediated oxidative/ER stress condition 1. Can J Physiol Pharmacol 2018; 97:441-456. [PMID: 30422673 DOI: 10.1139/cjpp-2018-0501] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Elevated homocysteine (Hcy), i.e., hyperhomocysteinemia (HHcy), causes skeletal muscle myopathy. Among many cellular and metabolic alterations caused by HHcy, oxidative and endoplasmic reticulum (ER) stress are considered the major ones; however, the precise molecular mechanism(s) in this process is unclear. Nevertheless, there is no treatment option available to treat HHcy-mediated muscle injury. Hydrogen sulfide (H2S) is increasingly recognized as a potent anti-oxidant, anti-apoptotic/necrotic/pyroptotic, and anti-inflammatory compound and also has been shown to improve angiogenesis during ischemic injury. Patients with CBS mutation produce less H2S, making them vulnerable to Hcy-mediated cellular damage. Many studies have reported bidirectional regulation of ER stress in apoptosis through JNK activation and concomitant attenuation of cell proliferation and protein synthesis via PI3K/AKT axis. Whether H2S mitigates these detrimental effects of HHcy on muscle remains unexplored. In this review, we discuss molecular mechanisms of HHcy-mediated oxidative/ER stress responses, apoptosis, angiogenesis, and atrophic changes in skeletal muscle and how H2S can restore skeletal muscle homeostasis during HHcy condition. This review also highlights the molecular mechanisms on how H2S could be developed as a clinically relevant therapeutic option for chronic conditions that are aggravated by HHcy.
Collapse
Affiliation(s)
- Avisek Majumder
- a Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA.,b Biochemistry and Molecular Genetics, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Mahavir Singh
- a Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA.,c Eye and Vision Science Laboratory, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Akash K George
- a Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA.,c Eye and Vision Science Laboratory, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Suresh C Tyagi
- a Department of Physiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
39
|
Majumder A, Singh M, George AK, Homme RP, Laha A, Tyagi SC. Remote ischemic conditioning as a cytoprotective strategy in vasculopathies during hyperhomocysteinemia: An emerging research perspective. J Cell Biochem 2018; 120:77-92. [PMID: 30272816 DOI: 10.1002/jcb.27603] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Accepted: 08/07/2018] [Indexed: 12/29/2022]
Abstract
Higher levels of nonprotein amino acid homocysteine (Hcy), that is, hyperhomocysteinemia (HHcy) (~5% of general population) has been associated with severe vasculopathies in different organs; however, precise molecular mechanism(s) as to how HHcy plays havoc with body's vascular networks are largely unknown. Interventional modalities have not proven beneficial to counter multifactorial HHcy's effects on the vascular system. An ancient Indian form of exercise called 'yoga' causes transient ischemia as a result of various body postures however the cellular mechanisms are not clear. We discuss a novel perspective wherein we argue that application of remote ischemic conditioning (RIC) could, in fact, deliver anticipated results to patients who are suffering from chronic vascular dysfunction due to HHcy. RIC is the mechanistic phenomenon whereby brief episodes of ischemia-reperfusion events are applied to distant tissues/organs; that could potentially offer a powerful tool in mitigating chronic lethal ischemia in target organs during HHcy condition via simultaneous reduction of inflammation, oxidative and endoplasmic reticulum stress, extracellular matrix remodeling, fibrosis, and angiogenesis. We opine that during ischemic conditioning our organs cross talk by releasing cellular messengers in the form of exosomes containing messenger RNAs, circular RNAs, anti-pyroptotic factors, protective cytokines like musclin, transcription factors, small molecules, anti-inflammatory, antiapoptotic factors, antioxidants, and vasoactive gases. All these could help mobilize the bone marrow-derived stem cells (having tissue healing properties) to target organs. In that context, we argue that RIC could certainly play a savior's role in an unfortunate ischemic or adverse event in people who have higher levels of the circulating Hcy in their systems.
Collapse
Affiliation(s)
- Avisek Majumder
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky.,Department of Biochemistry and Molecular Genetics, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Mahavir Singh
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky.,Eye and Vision Science Laboratory, University of Louisville, Louisville, Kentucky
| | - Akash K George
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky.,Eye and Vision Science Laboratory, University of Louisville, Louisville, Kentucky
| | - Rubens Petit Homme
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky.,Eye and Vision Science Laboratory, University of Louisville, Louisville, Kentucky
| | - Anwesha Laha
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| | - Suresh C Tyagi
- Department of Physiology, School of Medicine, University of Louisville, Louisville, Kentucky
| |
Collapse
|
40
|
Awad MA, Aldosari SR, Abid MR. Genetic Alterations in Oxidant and Anti-Oxidant Enzymes in the Vascular System. Front Cardiovasc Med 2018; 5:107. [PMID: 30140678 PMCID: PMC6095034 DOI: 10.3389/fcvm.2018.00107] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 07/16/2018] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular diseases (CVD) are one of the prime causes of mortality worldwide. Experimental animal models have become a valuable tool to investigate and further advance our knowledge on etiology, pathophysiology and intervention. They also provide a great opportunity to understand the contribution of different genes and effector molecules in the pathogenesis and development of diseases at the sub-cellular levels. High levels of reactive oxygen species (ROS) have been associated with the progression of CVD such as ischemic heart disease (IHD), myocardial infarction, hypertension, atherosclerosis, aortic aneurysm, aortic dissection and others. On the contrary, low levels of antioxidants were associated with exacerbated cardiovascular event. Major focus of this review is on vascular pathogenesis that leads to CVD, with special emphasis on the roles of oxidant/antioxidant enzymes in health and disease progression in vascular cells including vascular endothelium. The major oxidant enzymes that have been implicated with the progression of CVD include NADPH Oxidase, nitric oxide synthase, monoamine oxidase, and xanthine oxidoreductase. The major antioxidant enzymes that have been attributed to normalizing the levels of oxidative stress include superoxide dismutases, catalase and glutathione peroxidases (GPx), and thioredoxin. Cardiovascular phenotypes of major oxidants and antioxidants knockout and transgenic animal models are discussed here.
Collapse
Affiliation(s)
- Maan A Awad
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| | - Sarah R Aldosari
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| | - M Ruhul Abid
- Division of Cardiothoracic Surgery, Department of Surgery, Cardiovascular Research Center, Rhode Island Hospital, Brown University Alpert Medical School, Providence, RI, United States
| |
Collapse
|
41
|
Shinde AV, Su Y, Palanski BA, Fujikura K, Garcia MJ, Frangogiannis NG. Pharmacologic inhibition of the enzymatic effects of tissue transglutaminase reduces cardiac fibrosis and attenuates cardiomyocyte hypertrophy following pressure overload. J Mol Cell Cardiol 2018; 117:36-48. [PMID: 29481819 PMCID: PMC5892840 DOI: 10.1016/j.yjmcc.2018.02.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 01/26/2018] [Accepted: 02/22/2018] [Indexed: 12/11/2022]
Abstract
Tissue transglutaminase (tTG) is a multifunctional protein with a wide range of enzymatic and non-enzymatic functions. We have recently demonstrated that tTG expression is upregulated in the pressure-overloaded myocardium and exerts fibrogenic actions promoting diastolic dysfunction, while preventing chamber dilation. Our current investigation dissects the in vivo and in vitro roles of the enzymatic effects of tTG on fibrotic remodeling in pressure-overloaded myocardium. Using a mouse model of transverse aortic constriction, we demonstrated perivascular and interstitial tTG activation in the remodeling pressure-overloaded heart. tTG inhibition through administration of the selective small molecule tTG inhibitor ERW1041E attenuated left ventricular diastolic dysfunction and reduced cardiomyocyte hypertrophy and interstitial fibrosis in the pressure-overloaded heart, without affecting chamber dimensions and ejection fraction. In vivo, tTG inhibition markedly reduced myocardial collagen mRNA and protein levels and attenuated transcription of fibrosis-associated genes. In contrast, addition of exogenous recombinant tTG to fibroblast-populated collagen pads had no significant effects on collagen transcription, and instead increased synthesis of matrix metalloproteinase (MMP)3 and tissue inhibitor of metalloproteinases (TIMP)1 through transamidase-independent actions. However, enzymatic effects of matrix-bound tTG increased the thickness of pericellular collagen in fibroblast-populated pads. tTG exerts distinct enzymatic and non-enzymatic functions in the remodeling pressure-overloaded heart. The enzymatic effects of tTG are fibrogenic and promote diastolic dysfunction, but do not directly modulate the pro-fibrotic transcriptional program of fibroblasts. Targeting transamidase-dependent actions of tTG may be a promising therapeutic strategy in patients with heart failure and fibrosis-associated diastolic dysfunction.
Collapse
Affiliation(s)
- Arti V Shinde
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States
| | - Ya Su
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States
| | - Brad A Palanski
- Department of Chemistry, Stanford University, Stanford, CA, United States
| | - Kana Fujikura
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States
| | - Mario J Garcia
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States
| | - Nikolaos G Frangogiannis
- The Wilf Family Cardiovascular Research Institute, Department of Medicine (Cardiology), Albert Einstein College of Medicine, Bronx, NY, United States.
| |
Collapse
|
42
|
Zheng Z, Fan S, Zheng J, Huang W, Gasparetto C, Chao NJ, Hu J, Kang Y. Inhibition of thioredoxin activates mitophagy and overcomes adaptive bortezomib resistance in multiple myeloma. J Hematol Oncol 2018; 11:29. [PMID: 29482577 PMCID: PMC5828316 DOI: 10.1186/s13045-018-0575-7] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 02/12/2018] [Indexed: 11/25/2022] Open
Abstract
Background Although current chemotherapy using bortezomib (Velcade) against multiple myeloma in adults has achieved significant responses and even remission, a majority of patients will develop acquired resistance to bortezomib. Increased thioredoxin level has been reported to be associated with carcinogenesis; however, the role of thioredoxin in bortezomib drug resistance of myeloma remains unclear. Methods We generated several bortezomib-resistant myeloma cell lines by serially passaging with increased concentrations of bortezomib over a period of 1.5 years. Thioredoxin expression was measured by real-time PCR and western blot. Results The role of thioredoxin in the survival of bortezomib-resistant myeloma cells was determined by specific shRNA knockdown in vitro and in vivo. Thioredoxin inhibitor (PX12) was used to determine the effectiveness of thioredoxin inhibition in the treatment of bortezomib-resistant myeloma cells. The effect of thioredoxin inhibition on mitophagy induction was examined. The correlation of thioredoxin expression with patient overall survival was interrogated. Thioredoxin expression was significantly upregulated in bortezomib-resistant myeloma cells and the change correlated with the increase of bortezomib concentration. Thioredoxin gene knockdown using specific shRNA sensitized bortezomib-resistant myeloma cells to bortezomib efficiency in vitro and in vivo. Similarly, pharmacological inhibition with PX12 inhibited the growth of bortezomib-resistant myeloma cells and overcame bortezomib resistance in vitro and in vivo. Furthermore, inhibition of thioredoxin resulted in the activation of mitophagy and blockage of mitophagy prevented the effects of PX12 on bortezomib-resistant myeloma cells, indicating that mitophagy is the important molecular mechanism for the induction of cell death in bortezomib-resistant myeloma cells by PX12. Moreover, inhibition of thioredoxin resulted in downregulation of phosphorylated mTOR and ERK1/2. Finally, thioredoxin was overexpressed in primary myeloma cells isolated from bortezomib-resistant myeloma patients and overexpression of thioredoxin correlated with poor overall survival in patients with multiple myeloma. Conclusions Our findings demonstrated that increased thioredoxin plays a critical role in bortezomib resistance in multiple myeloma through mitophagy inactivation and increased mTOR and ERK1/2 phosphorylation. Thioredoxin provides a potential target for clinical therapeutics against multiple myeloma, particularly for bortezomib-resistant/refractory myeloma patients. Electronic supplementary material The online version of this article (10.1186/s13045-018-0575-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhihong Zheng
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China.,Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Shengjun Fan
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Jing Zheng
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China
| | - Wei Huang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Cristina Gasparetto
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Nelson J Chao
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA
| | - Jianda Hu
- Department of Hematology, Fujian Provincial Key Laboratory of Hematology, Fujian Medical University Union Hospital, 29 Xinquan Road, Fuzhou, Fujian, 350001, China.
| | - Yubin Kang
- Division of Hematologic Malignancies and Cellular Therapy, Duke University Medical Center, 3961, Durham, NC, 27710, USA.
| |
Collapse
|
43
|
Loss of myocardial protection against myocardial infarction in middle-aged transgenic mice overexpressing cardiac thioredoxin-1. Oncotarget 2017; 7:11889-98. [PMID: 26933812 PMCID: PMC4914256 DOI: 10.18632/oncotarget.7726] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2015] [Accepted: 01/30/2016] [Indexed: 11/25/2022] Open
Abstract
Thioredoxin-1 (Trx1) protects the heart from ischemia/reperfusion (I/R) injury. Given that the age at which the first episode of coronary disease takes place has considerably decreased, life at middle-aged (MA) emerges as a new field of study. The aim was determine whether infarct size, Trx1 expression and activity, Akt and GSK-3β were altered in young (Y) and MA mice overexpressing cardiac Trx1, and in a dominant negative (DN-Trx1) mutant of Trx1. Langendorff-perfused hearts were subjected to 30 minutes of ischemia and 120 minutes of reperfusion (R). We used 3 and 12 month-old male of wild type (WT), Trx1, and DN-Trx1. Trx1 overexpression reduced infarct size in young mice (WT-Y: 46.8±4.1% vs. Trx1-Y: 27.6±3.5%, p < 0.05). Trx1 activity was reduced by 52.3±3.2% (p < 0.05) in Trx1-MA, accompanied by an increase in nitration by 17.5±0.9%, although Trx1 expression in transgenic mice was similar between young and middle-aged. The expression of p-Akt and p-GSK-3β increased during reperfusion in Trx1-Y. DN-Trx1 mice showed neither reduction in infarct size nor Akt and GSK-3β phosphorylation. Our data suggest that the lack of protection in Trx1 middle-aged mice even with normal Trx1 expression may be associated to decreased Trx1 activity, increased nitration and inhibition of p-Akt and p-GSK-3β.
Collapse
|
44
|
Davies JMS, Cillard J, Friguet B, Cadenas E, Cadet J, Cayce R, Fishmann A, Liao D, Bulteau AL, Derbré F, Rébillard A, Burstein S, Hirsch E, Kloner RA, Jakowec M, Petzinger G, Sauce D, Sennlaub F, Limon I, Ursini F, Maiorino M, Economides C, Pike CJ, Cohen P, Salvayre AN, Halliday MR, Lundquist AJ, Jakowec NA, Mechta-Grigoriou F, Mericskay M, Mariani J, Li Z, Huang D, Grant E, Forman HJ, Finch CE, Sun PY, Pomatto LCD, Agbulut O, Warburton D, Neri C, Rouis M, Cillard P, Capeau J, Rosenbaum J, Davies KJA. The Oxygen Paradox, the French Paradox, and age-related diseases. GeroScience 2017; 39:499-550. [PMID: 29270905 PMCID: PMC5745211 DOI: 10.1007/s11357-017-0002-y] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2017] [Accepted: 11/09/2017] [Indexed: 02/06/2023] Open
Abstract
A paradox is a seemingly absurd or impossible concept, proposition, or theory that is often difficult to understand or explain, sometimes apparently self-contradictory, and yet ultimately correct or true. How is it possible, for example, that oxygen "a toxic environmental poison" could be also indispensable for life (Beckman and Ames Physiol Rev 78(2):547-81, 1998; Stadtman and Berlett Chem Res Toxicol 10(5):485-94, 1997)?: the so-called Oxygen Paradox (Davies and Ursini 1995; Davies Biochem Soc Symp 61:1-31, 1995). How can French people apparently disregard the rule that high dietary intakes of cholesterol and saturated fats (e.g., cheese and paté) will result in an early death from cardiovascular diseases (Renaud and de Lorgeril Lancet 339(8808):1523-6, 1992; Catalgol et al. Front Pharmacol 3:141, 2012; Eisenberg et al. Nat Med 22(12):1428-1438, 2016)?: the so-called, French Paradox. Doubtless, the truth is not a duality and epistemological bias probably generates apparently self-contradictory conclusions. Perhaps nowhere in biology are there so many apparently contradictory views, and even experimental results, affecting human physiology and pathology as in the fields of free radicals and oxidative stress, antioxidants, foods and drinks, and dietary recommendations; this is particularly true when issues such as disease-susceptibility or avoidance, "healthspan," "lifespan," and ageing are involved. Consider, for example, the apparently paradoxical observation that treatment with low doses of a substance that is toxic at high concentrations may actually induce transient adaptations that protect against a subsequent exposure to the same (or similar) toxin. This particular paradox is now mechanistically explained as "Adaptive Homeostasis" (Davies Mol Asp Med 49:1-7, 2016; Pomatto et al. 2017a; Lomeli et al. Clin Sci (Lond) 131(21):2573-2599, 2017; Pomatto and Davies 2017); the non-damaging process by which an apparent toxicant can activate biological signal transduction pathways to increase expression of protective genes, by mechanisms that are completely different from those by which the same agent induces toxicity at high concentrations. In this review, we explore the influences and effects of paradoxes such as the Oxygen Paradox and the French Paradox on the etiology, progression, and outcomes of many of the major human age-related diseases, as well as the basic biological phenomenon of ageing itself.
Collapse
Affiliation(s)
- Joanna M S Davies
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Josiane Cillard
- Lab de Biologie Cellulaire et Végétale, Faculté de Pharmacie, Université de Rennes, 35043, Rennes Cedex, France
| | - Bertrand Friguet
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - Enrique Cadenas
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- School of Pharmacy, University of Southern California, Los Angeles, CA, 90089-9121, USA
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Jean Cadet
- Département de Médecine nucléaire et Radiobiologie, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, J1H 5N4, Canada
| | - Rachael Cayce
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Andrew Fishmann
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - David Liao
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Anne-Laure Bulteau
- Institut de Génomique Fonctionnelle de Lyon,ENS de Lyon, CNRS, 69364, Lyon Cedex 07, France
| | - Frédéric Derbré
- Laboratory for Movement, Sport and Health Sciences-EA 1274, M2S, Université de Rennes 2-ENS, Bruz, 35170, Rennes, France
| | - Amélie Rébillard
- Laboratory for Movement, Sport and Health Sciences-EA 1274, M2S, Université de Rennes 2-ENS, Bruz, 35170, Rennes, France
| | - Steven Burstein
- The Medical Group, Internal Medicine, Rheumatology & Osteoporosis, Dermatology, Pulmonology, Ophthalmology, and Cardiology; the Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Etienne Hirsch
- INSERM UMR 1127-CNRS UMR 7225, Institut du cerveau et de la moelle épinière-ICM Thérapeutique Expérimentale de la Maladie de Parkinson, Université Pierre et Marie Curie, 75651, Paris Cedex 13, France
| | - Robert A Kloner
- Huntington Medical Research Institutes, Pasadena, CA, 91105, USA
| | - Michael Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Giselle Petzinger
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Delphine Sauce
- Chronic infections and Immune ageing, INSERM U1135, Hopital Pitie-Salpetriere, Pierre et Marie Curie University, 75013, Paris, France
| | | | - Isabelle Limon
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Fulvio Ursini
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Matilde Maiorino
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Christina Economides
- Los Angeles Cardiology Associates, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Christian J Pike
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Neurobiology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Pinchas Cohen
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Anne Negre Salvayre
- Lipid peroxidation, Signalling and Vascular Diseases INSERM U1048, 31432, Toulouse Cedex 4, France
| | - Matthew R Halliday
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Adam J Lundquist
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | - Nicolaus A Jakowec
- Department of Neurology, Keck School of Medicine, University of Southern California, Los Angeles, CA, 90033, USA
| | | | - Mathias Mericskay
- Laboratoire de Signalisation et Physiopathologie Cardiovasculaire-Inserm UMR-S 1180, Faculté de Pharmacie, Université Paris-Sud, 92296 Châtenay-Malabry, Paris, France
| | - Jean Mariani
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Zhenlin Li
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - David Huang
- Department of Radiation Oncology, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Ellsworth Grant
- Department of Oncology & Hematology, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
| | - Henry J Forman
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Caleb E Finch
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Los Angeles Cardiology Associates, Hospital of the Good Samaritan, Los Angeles, CA, 90017, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Patrick Y Sun
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Laura C D Pomatto
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA
| | - Onnik Agbulut
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - David Warburton
- Children's Hospital of Los Angeles, Developmental Biology, Regenerative Medicine and Stem Cell Therapeutics program and the Center for Environmental Impact on Global Health Across the Lifespan at The Saban Research Institute, Los Angeles, CA, 90027, USA
- Department of Pediatrics, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA
| | - Christian Neri
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
| | - Mustapha Rouis
- Institut de Biologie Paris-Seine (IBPS), UMR CNRS 8256, Biological Adaptation and Ageing, Sorbonne Universités, UPMC Univ Paris 06, 75005, Paris, France
- INSERM ERL U1164, 75005, Paris, France
| | - Pierre Cillard
- Lab de Biologie Cellulaire et Végétale, Faculté de Pharmacie, Université de Rennes, 35043, Rennes Cedex, France
| | - Jacqueline Capeau
- DR Saint-Antoine UMR_S938, UPMC, Inserm Faculté de Médecine, Université Pierre et Marie Curie, 75012, Paris, France
| | - Jean Rosenbaum
- Scientific Service of the Embassy of France in the USA, Consulate General of France in Los Angeles, Los Angeles, CA, 90025, USA
| | - Kelvin J A Davies
- Leonard Davis School of Gerontology of the Ethel Percy Andrus Gerontology Center, University of Southern California, Los Angeles, CA, 90089-0191, USA.
- Department of Biochemistry & Molecular Medicine, Keck School of Medicine of USC, University of Southern California, Los Angeles, CA, 90033, USA.
- Division of Molecular & Computational Biology, Department of Biological Sciences of the Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA, 90089-0191, USA.
| |
Collapse
|
45
|
Su H, Pistolozzi M, Shi X, Sun X, Tan W. Alterations in NO/ROS ratio and expression of Trx1 and Prdx2 in isoproterenol-induced cardiac hypertrophy. Acta Biochim Biophys Sin (Shanghai) 2017; 49:1022-1028. [PMID: 29036266 DOI: 10.1093/abbs/gmx102] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Indexed: 12/27/2022] Open
Abstract
The development of cardiac hypertrophy is a complicated process, which undergoes a transition from compensatory hypertrophy to heart failure, and the identification of new biomarkers and targets for this disease is greatly needed. Here we investigated the development of isoproterenol (ISO)-induced cardiac hypertrophy in an in vitro experimental model. After the induction of hypertrophy with ISO treatment in H9c2 cells, cell surface area, cell viability, cellular reactive oxygen species (ROS), and nitric oxide (NO) levels were tested. Our data showed that the cell viability, mitochondrial membrane potential, and NO/ROS balance varied during the development of cardiac hypertrophy in H9c2 cells. It was also found that the expression of thioredoxin1 (Trx1) and peroxiredoxin2 (Prdx2) was decreased during the cardiac hypertrophy of H9c2 cells. These results suggest a critical role for Trx1 and Prdx2 in the cardiac hypertrophy of H9c2 cells and in the transition from compensated hypertrophy to de-compensated hypertrophy in H9c2 cells, and our findings may have important implications for the management of this disease.
Collapse
Affiliation(s)
- Hao Su
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Marco Pistolozzi
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Xingjuan Shi
- Key Laboratory of Developmental Genes and Human Disease, Institute of Life Sciences, Southeast University, Nanjing 210096, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
- School of Bioscience and Bioengineering, South China University of Technology, Guangzhou 510006, China
| | - Wen Tan
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| |
Collapse
|
46
|
Oka SI, Hirata T, Suzuki W, Naito D, Chen Y, Chin A, Yaginuma H, Saito T, Nagarajan N, Zhai P, Bhat S, Schesing K, Shao D, Hirabayashi Y, Yodoi J, Sciarretta S, Sadoshima J. Thioredoxin-1 maintains mechanistic target of rapamycin (mTOR) function during oxidative stress in cardiomyocytes. J Biol Chem 2017; 292:18988-19000. [PMID: 28939765 DOI: 10.1074/jbc.m117.807735] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 09/06/2017] [Indexed: 12/19/2022] Open
Abstract
Thioredoxin 1 (Trx1) is a 12-kDa oxidoreductase that catalyzes thiol-disulfide exchange reactions to reduce proteins with disulfide bonds. As such, Trx1 helps protect the heart against stresses, such as ischemia and pressure overload. Mechanistic target of rapamycin (mTOR) is a serine/threonine kinase that regulates cell growth, metabolism, and survival. We have shown previously that mTOR activity is increased in response to myocardial ischemia-reperfusion injury. However, whether Trx1 interacts with mTOR to preserve heart function remains unknown. Using a substrate-trapping mutant of Trx1 (Trx1C35S), we show here that mTOR is a direct interacting partner of Trx1 in the heart. In response to H2O2 treatment in cardiomyocytes, mTOR exhibited a high molecular weight shift in non-reducing SDS-PAGE in a 2-mercaptoethanol-sensitive manner, suggesting that mTOR is oxidized and forms disulfide bonds with itself or other proteins. The mTOR oxidation was accompanied by reduced phosphorylation of endogenous substrates, such as S6 kinase (S6K) and 4E-binding protein 1 (4E-BP1) in cardiomyocytes. Immune complex kinase assays disclosed that H2O2 treatment diminished mTOR kinase activity, indicating that mTOR is inhibited by oxidation. Of note, Trx1 overexpression attenuated both H2O2-mediated mTOR oxidation and inhibition, whereas Trx1 knockdown increased mTOR oxidation and inhibition. Moreover, Trx1 normalized H2O2-induced down-regulation of metabolic genes and stimulation of cell death, and an mTOR inhibitor abolished Trx1-mediated rescue of gene expression. H2O2-induced oxidation and inhibition of mTOR were attenuated when Cys-1483 of mTOR was mutated to phenylalanine. These results suggest that Trx1 protects cardiomyocytes against stress by reducing mTOR at Cys-1483, thereby preserving the activity of mTOR and inhibiting cell death.
Collapse
Affiliation(s)
- Shin-Ichi Oka
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Tsuyoshi Hirata
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Wataru Suzuki
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Daichi Naito
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Yanbin Chen
- the Department of Respiratory Medicine, First Affiliated Hospital of Soochow University, 899 Pinghai Road, Suzhou 215000, China
| | - Adave Chin
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Hiroaki Yaginuma
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Toshiro Saito
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Narayani Nagarajan
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Peiyong Zhai
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Santosh Bhat
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Kevin Schesing
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Dan Shao
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101
| | - Yoko Hirabayashi
- the Division of Cellular and Molecular Toxicology, Center for Biological Safety and Research, National Institute of Health Sciences, Tokyo 158-8501, Japan
| | - Junji Yodoi
- the Department of Biological Responses, Laboratory of Infection and Prevention, Institute for Virus Research, Kyoto University, Sakyo-ku, Kyoto 606-8397, Japan, and
| | - Sebastiano Sciarretta
- the Department of Medical-Surgical Science and Biotechnologies, University of Rome, Latina 04100, Italy
| | - Junichi Sadoshima
- From the Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, New Jersey 07101,
| |
Collapse
|
47
|
Yang J, Hamid S, Liu Q, Cai J, Xu S, Zhang Z. Gene expression of selenoproteins can be regulated by thioredoxin(Txn) silence in chicken cardiomyocytes. J Inorg Biochem 2017; 177:118-126. [PMID: 28957736 DOI: 10.1016/j.jinorgbio.2017.08.027] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 08/28/2017] [Accepted: 08/30/2017] [Indexed: 02/06/2023]
Abstract
Thioredoxin (Txn) system is the most crucial antioxidant defense mechanism in myocardium. The aim of this study was to clarify the effect of Txn low expression on 25 selenoproteins in chicken cardiomyocytes. We developed a Se-deficient model (0.033mg/kg) and Txn knock down cardiomyocytes model (siRNA) studies. Western Blot, Quantitative Real-time PCR (qPCR) were performed, and correlation analysis, heat map were used for further analysis. Both low expression of Txn models are significantly decreased (P<0.05) the mRNA levels of Deiodinase 1, 2 (Dio 1, 2), Glutathione Peroxidase 1, 2, 3, 4 (Gpx 1, 2, 3, 4), Thioredoxin Reductase 1, 2, 3 (TR 1, 2, 3), Selenoprotein t (Selt), Selenoprotein w (Selw), Selenoprotein k (Selk), selenoprotein x1 (Sepx1), and significantly increased (P<0.05) the mRNA levels of the rest of selenoproteins. Correlation analysis showed that Deiodinase 3 (Dio 3), Selenoprotein m (Selm), 15-kDa Selenoprotein (Selp15), Selenoprotein h (Selh), Selenoprotein u (Selu), Selenoprotein i (Seli), Selenoprotein n (Seln), Selenoprotein p1 (Sepp1), Selenoprotein o (Selo), Selenoprotein s (Sels), Selenoprotein synthetase 2 (Sels2) and Selenoprotein p (Selp) had a negative correlation with Txn, while the rest of selenoproteins had a positive correlation with Txn. Combined in vivo and in vitro we can know that hamper Txn expression can inhibit Gpx 1, 2, 3, 4, TR 1, 2, 3, Dio 1, 2, Selt, Selw, Selk, Sepx1, meanwhile, over expression the rest of selenoproteins. In conclusion, the different selenoproteins possess and exhibit distinct responses to silence of Txn in chicken cardiomyocytes.
Collapse
Affiliation(s)
- Jie Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Sattar Hamid
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Qi Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Jingzeng Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China
| | - Shiwen Xu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, PR China.
| |
Collapse
|
48
|
Kanaan GN, Harper ME. Cellular redox dysfunction in the development of cardiovascular diseases. Biochim Biophys Acta Gen Subj 2017; 1861:2822-2829. [PMID: 28778485 DOI: 10.1016/j.bbagen.2017.07.027] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/21/2017] [Accepted: 07/30/2017] [Indexed: 12/22/2022]
Abstract
To meet its exceptionally high energy demands, the heart relies largely on fatty acid oxidation, which then drives the oxidative phosphorylation system in mitochondria. Each day, this system produces about 6kg of ATP to sustain heart function. Fatty acid oxidation is sometimes associated with high rates of mitochondrial reactive oxygen species (ROS) production. By definition, ROS are singlet electron intermediates formed during the partial reduction of oxygen to water and they include radical and non-radical intermediates like superoxide, hydrogen peroxide and hydroxyl radical. Superoxide can also interact with nitric oxide to produce peroxynitrite that in turn can give rise to other radical or non-radical reactive nitrogen species (RNS) like nitrogen dioxide, dinitrogen trioxide and others. While mitochondrial and cellular functions can be impaired by ROS if they accumulate, under normal physiological conditions ROS are important signaling molecules in the cardiovascular system. A fine balance between ROS production and antioxidant systems, including glutathione redox, is essential in the heart; otherwise the ensuing damage can contribute to pathogenic processes, which can culminate in endothelial dysfunction, atherosclerosis, hypertension, cardiac hypertrophy, arrhythmias, myocardial ischemia/reperfusion damage, and heart failure. Here we provide a succinct review of recent findings.
Collapse
Affiliation(s)
- Georges N Kanaan
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Ottawa Institute of Systems Biology, Faculty of Medicine, University of Ottawa, Ottawa, Canada.
| |
Collapse
|
49
|
Nagarajan N, Oka S, Sadoshima J. Modulation of signaling mechanisms in the heart by thioredoxin 1. Free Radic Biol Med 2017; 109:125-131. [PMID: 27993729 PMCID: PMC5462876 DOI: 10.1016/j.freeradbiomed.2016.12.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Accepted: 12/14/2016] [Indexed: 01/15/2023]
Abstract
Myocardial ischemia/reperfusion and heart failure are the major cardiac conditions in which an imbalance between oxidative stress and anti-oxidant mechanisms is observed. The myocardium has endogenous reducing mechanisms, including the thioredoxin (Trx) and glutathione systems, that act to scavenge reactive oxygen species (ROS) and reduce oxidized proteins. The Trx system consists of Trx, Trx reductase (TrxR), and an electron donor, NADPH, where Trx is maintained in a reduced state in the presence of TrxR and NADPH. Trx1, a major isoform of Trx, is abundantly expressed in the heart and exerts its oxidoreductase activity through conserved Cys32 and Cys35, reducing oxidized proteins through thiol disulfide exchange reactions. In this review, we will focus on molecular targets of Trx1 in the heart, including transcription factors, microRNAs, histone deactylases, and protein kinases. We will then discuss how Trx1 regulates the functions of its targets, thereby affecting the extent of myocardial injury caused by myocardial ischemia/reperfusion and the progression of heart failure.
Collapse
Affiliation(s)
- Narayani Nagarajan
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G609, Newark, NJ 07103, USA
| | - Shinichi Oka
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G609, Newark, NJ 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G609, Newark, NJ 07103, USA.
| |
Collapse
|
50
|
Bubb KJ, Birgisdottir AB, Tang O, Hansen T, Figtree GA. Redox modification of caveolar proteins in the cardiovascular system- role in cellular signalling and disease. Free Radic Biol Med 2017; 109:61-74. [PMID: 28188926 DOI: 10.1016/j.freeradbiomed.2017.02.012] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 01/18/2017] [Accepted: 02/05/2017] [Indexed: 02/07/2023]
Abstract
Rapid and coordinated release of a variety of reactive oxygen species (ROS) such as superoxide (O2.-), hydrogen peroxide (H2O2) and peroxynitrite, in specific microdomains, play a crucial role in cell signalling in the cardiovascular system. These reactions are mediated by reversible and functional modifications of a wide variety of key proteins. Dysregulation of this oxidative signalling occurs in almost all forms of cardiovascular disease (CVD), including at the very early phases. Despite the heavily publicized failure of "antioxidants" to improve CVD progression, pharmacotherapies such as those targeting the renin-angiotensin system, or statins, exert at least part of their large clinical benefit via modulating cellular redox signalling. Over 250 proteins, including receptors, ion channels and pumps, and signalling proteins are found in the caveolae. An increasing proportion of these are being recognized as redox regulated-proteins, that reside in the immediate vicinity of the two major cellular sources of ROS, nicotinamide adenine dinucleotide phosphate oxidase (Nox) and uncoupled endothelial nitric oxide synthase (eNOS). This review focuses on what is known about redox signalling within the caveolae, as well as endogenous protective mechanisms utilized by the cell, and new approaches to targeting dysregulated redox signalling in the caveolae as a therapeutic strategy in CVD.
Collapse
Affiliation(s)
- Kristen J Bubb
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Asa Birna Birgisdottir
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia; Department of Cardiothoracic and Vascular Surgery, Heart and Lung Clinic, University Hospital of North Norway, Tromsø, Norway
| | - Owen Tang
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Thomas Hansen
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia
| | - Gemma A Figtree
- Kolling Institute of Medical Research, University of Sydney and Cardiology Department, Royal North Shore Hospital, St Leonards, NSW 2065, Australia.
| |
Collapse
|