1
|
Grayson C, Chalifoux O, Russo MDST, Avizonis DZ, Sterman S, Faerman B, Koufos O, Agellon LB, Mailloux RJ. Ablating the glutaredoxin-2 (Glrx2) gene protects male mice against non-alcoholic fatty liver disease (NAFLD) by limiting oxidative distress. Free Radic Biol Med 2024; 224:660-677. [PMID: 39278573 DOI: 10.1016/j.freeradbiomed.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/22/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
In the present study, we investigated the consequences of deleting the glutaredoxin-2 gene (Glrx2-/-) on the development of non-alcoholic fatty liver disease (NAFLD) in male and female C57BL6N mice fed a control (CD) or high-fat diet (HFD). We report that the HFD induced a significant increase in body mass in the wild-type (Wt) and Glrx2-/- male, but not female, mice, which was associated with the hypertrophying of the abdominal fat. Interestingly, while the Wt male mice fed the HFD developed NAFLD, the deletion of the Glrx2 gene mitigated vesicle formation, intrahepatic lipid accumulation, and fibrosis in the males. The protective effect associated with ablating the Glrx2 gene in male mice was due to enhancement of mitochondrial redox buffering capacity. Specifically, liver mitochondria from male Glrx2-/- fed a CD or HFD produced significantly less hydrogen peroxide (mtH2O2), had lower malondialdehyde levels, greater activities for glutathione peroxidase and thioredoxin reductase, and less protein glutathione mixed disulfides (PSSG) when compared to the Wt male mice fed the HFD. These effects correlated with the S-glutathionylation of α-ketoglutarate dehydrogenase (KGDH), a potent mtH2O2 source and key redox sensor in hepatic mitochondria. In comparison to the male mice, both Wt and Glrx2-/- female mice displayed almost complete resistance to HFD-induced body mass increases and the development of NAFLD, which was attributed to the superior redox buffering capacity of the liver mitochondria. Together, our findings show that modulation of mitochondrial S-glutathionylation signaling through Glrx2 augments resistance of male mice towards the development of NAFLD through preservation of mitochondrial redox buffering capacity. Additionally, our findings demonstrate the sex dimorphisms associated with the manifestation of NAFLD is related to the superior redox buffering capacity and modulation of the S-glutathionylome in hepatic mitochondria from female mice.
Collapse
Affiliation(s)
- Cathryn Grayson
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Olivia Chalifoux
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Mariana De Sa Tavares Russo
- Goodman Cancer Institute, McGill University, Qc, H3A 1A3, Montréal, Québec, Canada; Department of Medicine, McGill University, Qc, H3A 1A3, Montréal, Québec, Canada
| | - Daina Zofija Avizonis
- Goodman Cancer Institute, McGill University, Qc, H3A 1A3, Montréal, Québec, Canada; Department of Medicine, McGill University, Qc, H3A 1A3, Montréal, Québec, Canada
| | - Samantha Sterman
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Ben Faerman
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Olivia Koufos
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Luis B Agellon
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada
| | - Ryan J Mailloux
- School of Human Nutrition, Faculty of Agricultural and Environmental Science, McGill University, Ste Anne de Bellevue, Québec, Canada.
| |
Collapse
|
2
|
Zhou M, Hanschmann EM, Römer A, Linn T, Petry SF. The significance of glutaredoxins for diabetes mellitus and its complications. Redox Biol 2024; 71:103043. [PMID: 38377787 PMCID: PMC10891345 DOI: 10.1016/j.redox.2024.103043] [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: 12/09/2023] [Accepted: 01/13/2024] [Indexed: 02/22/2024] Open
Abstract
Diabetes mellitus is a non-communicable metabolic disease hallmarked by chronic hyperglycemia caused by beta-cell failure. Diabetic complications affect the vasculature and result in macro- and microangiopathies, which account for a significantly increased morbidity and mortality. The rising incidence and prevalence of diabetes is a major global health burden. There are no feasible strategies for beta-cell preservation available in daily clinical practice. Therefore, patients rely on antidiabetic drugs or the application of exogenous insulin. Glutaredoxins (Grxs) are ubiquitously expressed and highly conserved members of the thioredoxin family of proteins. They have specific functions in redox-mediated signal transduction, iron homeostasis and biosynthesis of iron-sulfur (FeS) proteins, and the regulation of cell proliferation, survival, and function. The involvement of Grxs in chronic diseases has been a topic of research for several decades, suggesting them as therapeutic targets. Little is known about their role in diabetes and its complications. Therefore, this review summarizes the available literature on the significance of Grxs in diabetes and its complications. In conclusion, Grxs are differentially expressed in the endocrine pancreas and in tissues affected by diabetic complications, such as the heart, the kidneys, the eye, and the vasculature. They are involved in several pathways essential for insulin signaling, metabolic inflammation, glucose and fatty acid uptake and processing, cell survival, and iron and mitochondrial metabolism. Most studies describe significant changes in glutaredoxin expression and/or activity in response to the diabetic metabolism. In general, mitigated levels of Grxs are associated with oxidative distress, cell damage, and even cell death. The induced overexpression is considered a potential part of the cellular stress-response, counteracting oxidative distress and exerting beneficial impact on cell function such as insulin secretion, cytokine expression, and enzyme activity.
Collapse
Affiliation(s)
- Mengmeng Zhou
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Eva-Maria Hanschmann
- Experimental and Translational Research, Department of Otorhinolaryngology, University Hospital Essen, Essen, Germany
| | - Axel Römer
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Thomas Linn
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany
| | - Sebastian Friedrich Petry
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, Giessen, Germany.
| |
Collapse
|
3
|
Liu Y, Gong J, Wang Q, Wei N, Zhao L, Wu Z. Activation of the Nrf2/HO-1 axis by glutaredoxin 2 overexpression antagonizes vascular endothelial cell oxidative injury and inflammation under LPS exposure. Cytotechnology 2024; 76:167-178. [PMID: 38495299 PMCID: PMC10940561 DOI: 10.1007/s10616-023-00606-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 11/03/2023] [Indexed: 03/19/2024] Open
Abstract
Atherosclerosis constitutes a proverbial pathogenic mechanism for cardio-cerebrovascular disease that accounts for the most common cause of disability and morbidity for human health worldwide. Endothelial dysfunction and inflammation are the key contributors to the progression of atherosclerosis. Glutaredoxin 2 (GLRX2) is abundantly existed in various tissues and possesses a range of pleiotropic efficacy including anti-oxidative and anti-inflammatory responses. However, its role in atherosclerosis is still undefined. Here, down-regulation of GLRX2 was validated in lipopolysaccha (LPS)-induced vascular endothelial cells (HUVECs). Moreover, elevation of GLRX2 reversed the inhibition of cell viability in LPS-treated HUVECs and decreased LPS-induced increases in cell apoptosis and caspase-3 activity. Additionally, enhancement of GLRX2 expression antagonized oxidative stress in HUVECs under LPS exposure by inhibiting ROS, lactate dehydrogenase and malondialdehyde production and increased activity of anti-oxidative stress superoxide dismutase. Notably, GLRX2 abrogated LPS-evoked transcripts and releases of pro-inflammatory cytokine (TNF-α, IL-6, and IL-1β), chemokine MCP-1 and adhesion molecule ICAM-1 expression. Furthermore, the activation of Nrf2/HO-1 signaling was demonstrated in LPS-stimulated HUVECs. Importantly, blockage of the Nrf2 pathway counteracted the protective roles of GLRX2 in LPS-triggered endothelial cell injury, oxidative stress and inflammatory response. Thus, these data reveal that GLRX2 may alleviate the progression of atherosclerosis by regulating vascular endothelial dysfunction and inflammation via the activation of the Nrf2 signaling, supporting a promising therapeutic approach for atherosclerosis and its complications. Supplementary Information The online version contains supplementary material available at 10.1007/s10616-023-00606-x.
Collapse
Affiliation(s)
- Yuna Liu
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Yongding Road East Street, Beijing, 100039 People’s Republic of China
| | - Jinlin Gong
- Department of Medical Technology Support, Jingxi Medical District, Chinese PLA General Hospital, Beijing, 100097 People’s Republic of China
| | - Qing Wang
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Yongding Road East Street, Beijing, 100039 People’s Republic of China
| | - Na Wei
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Yongding Road East Street, Beijing, 100039 People’s Republic of China
| | - Lei Zhao
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Yongding Road East Street, Beijing, 100039 People’s Republic of China
| | - Zhenan Wu
- Department of Clinical Laboratory, Beijing Hospital of Integrated Traditional Chinese and Western Medicine, No. 3 Yongding Road East Street, Beijing, 100039 People’s Republic of China
| |
Collapse
|
4
|
Mouratidou C, Pavlidis ET, Katsanos G, Kotoulas SC, Mouloudi E, Tsoulfas G, Galanis IN, Pavlidis TE. Hepatic ischemia-reperfusion syndrome and its effect on the cardiovascular system: The role of treprostinil, a synthetic prostacyclin analog. World J Gastrointest Surg 2023; 15:1858-1870. [PMID: 37901735 PMCID: PMC10600776 DOI: 10.4240/wjgs.v15.i9.1858] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/20/2023] [Accepted: 07/25/2023] [Indexed: 09/21/2023] Open
Abstract
Hepatic ischemia-reperfusion syndrome has been the subject of intensive study and experimentation in recent decades since it is responsible for the outcome of several clinical entities, such as major hepatic resections and liver transplantation. In addition to the organ's post reperfusion injury, this syndrome appears to play a central role in the dysfunction of distant tissues and systems. Thus, continuous research should be directed toward finding effective therapeutic options to improve the outcome and reduce the postoperative morbidity and mortality rates. Treprostinil is a synthetic analog of prostaglandin I2, and its experimental administration has shown encouraging results. It has already been approved by the Food and Drug Administration in the United States for pulmonary arterial hypertension and has been used in liver transplantation, where preliminary encouraging results showed its safety and feasibility by using continuous intravenous administration at a dose of 5 ng/kg/min. Treprostinil improves renal and hepatic function, diminishes hepatic oxidative stress and lipid peroxidation, reduces hepatictoll-like receptor 9 and inflammation, inhibits hepatic apoptosis and restores hepatic adenosine triphosphate (ATP) levels and ATP synthases, which is necessary for functional maintenance of mitochondria. Treprostinil exhibits vasodilatory properties and antiplatelet activity and regulates proinflammatory cytokines; therefore, it can potentially minimize ischemia-reperfusion injury. Additionally, it may have beneficial effects on cardiovascular parameters, and much current research interest is concentrated on this compound.
Collapse
Affiliation(s)
| | - Efstathios T Pavlidis
- 2nd Propedeutic Department of Surgery, Hippokration General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| | - Georgios Katsanos
- Department of Transplantation, Hippokration General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| | | | - Eleni Mouloudi
- Intensive Care Unit, Hippokration General Hospital, Thessaloniki 54642, Greece
| | - Georgios Tsoulfas
- Department of Transplantation, Hippokration General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| | - Ioannis N Galanis
- 2nd Propedeutic Department of Surgery, Hippokration General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| | - Theodoros E Pavlidis
- 2nd Propedeutic Department of Surgery, Hippokration General Hospital, School of Medicine, Aristotle University of Thessaloniki, Thessaloniki 54642, Greece
| |
Collapse
|
5
|
Regulation of Mitochondrial Hydrogen Peroxide Availability by Protein S-glutathionylation. Cells 2022; 12:cells12010107. [PMID: 36611901 PMCID: PMC9818751 DOI: 10.3390/cells12010107] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND It has been four decades since protein S-glutathionylation was proposed to serve as a regulator of cell metabolism. Since then, this redox-sensitive covalent modification has been identified as a cell-wide signaling platform required for embryonic development and regulation of many physiological functions. SCOPE OF THE REVIEW Mitochondria use hydrogen peroxide (H2O2) as a second messenger, but its availability must be controlled to prevent oxidative distress and promote changes in cell behavior in response to stimuli. Experimental data favor the function of protein S-glutathionylation as a feedback loop for the inhibition of mitochondrial H2O2 production. MAJOR CONCLUSIONS The glutathione pool redox state is linked to the availability of H2O2, making glutathionylation an ideal mechanism for preventing oxidative distress whilst playing a part in desensitizing mitochondrial redox signals. GENERAL SIGNIFICANCE The biological significance of glutathionylation is rooted in redox status communication. The present review critically evaluates the experimental evidence supporting its role in negating mitochondrial H2O2 production for cell signaling and prevention of electrophilic stress.
Collapse
|
6
|
Vrettou S, Wirth B. S-Glutathionylation and S-Nitrosylation in Mitochondria: Focus on Homeostasis and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:15849. [PMID: 36555492 PMCID: PMC9779533 DOI: 10.3390/ijms232415849] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/24/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Redox post-translational modifications are derived from fluctuations in the redox potential and modulate protein function, localization, activity and structure. Amongst the oxidative reversible modifications, the S-glutathionylation of proteins was the first to be characterized as a post-translational modification, which primarily protects proteins from irreversible oxidation. However, a growing body of evidence suggests that S-glutathionylation plays a key role in core cell processes, particularly in mitochondria, which are the main source of reactive oxygen species. S-nitrosylation, another post-translational modification, was identified >150 years ago, but it was re-introduced as a prototype cell-signaling mechanism only recently, one that tightly regulates core processes within the cell’s sub-compartments, especially in mitochondria. S-glutathionylation and S-nitrosylation are modulated by fluctuations in reactive oxygen and nitrogen species and, in turn, orchestrate mitochondrial bioenergetics machinery, morphology, nutrients metabolism and apoptosis. In many neurodegenerative disorders, mitochondria dysfunction and oxidative/nitrosative stresses trigger or exacerbate their pathologies. Despite the substantial amount of research for most of these disorders, there are no successful treatments, while antioxidant supplementation failed in the majority of clinical trials. Herein, we discuss how S-glutathionylation and S-nitrosylation interfere in mitochondrial homeostasis and how the deregulation of these modifications is associated with Alzheimer’s, Parkinson’s, amyotrophic lateral sclerosis and Friedreich’s ataxia.
Collapse
Affiliation(s)
- Sofia Vrettou
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
7
|
Wang K, Hirschenson J, Moore A, Mailloux RJ. Conditions Conducive to the Glutathionylation of Complex I Subunit NDUFS1 Augment ROS Production following the Oxidation of Ubiquinone Linked Substrates, Glycerol-3-Phosphate and Proline. Antioxidants (Basel) 2022; 11:2043. [PMID: 36290766 PMCID: PMC9598259 DOI: 10.3390/antiox11102043] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/05/2022] [Accepted: 10/14/2022] [Indexed: 08/15/2023] Open
Abstract
Mitochondrial complex I can produce large quantities of reactive oxygen species (ROS) by reverse electron transfer (RET) from the ubiquinone (UQ) pool. Glutathionylation of complex I does induce increased mitochondrial superoxide/hydrogen peroxide (O2●-/H2O2) production, but the source of this ROS has not been identified. Here, we interrogated the glutathionylation of complex I subunit NDUFS1 and examined if its modification can result in increased ROS production during RET from the UQ pool. We also assessed glycerol-3-phosphate dehydrogenase (GPD) and proline dehydrogenase (PRODH) glutathionylation since both flavoproteins have measurable rates for ROS production as well. Induction of glutathionylation with disulfiram induced a significant increase in O2●-/H2O2 production during glycerol-3-phosphate (G3P) and proline (Pro) oxidation. Treatment of mitochondria with inhibitors for complex I (rotenone and S1QEL), complex III (myxothiazol and S3QEL), glycerol-3-phosphate dehydrogenase (iGP), and proline dehydrogenase (TFA) confirmed that the sites for this increase were complexes I and III, respectively. Treatment of liver mitochondria with disulfiram (50-1000 nM) did not induce GPD or PRODH glutathionylation, nor did it affect their activities, even though disulfiram dose-dependently increased the total number of protein glutathione mixed disulfides (PSSG). Immunocapture of complex I showed disulfiram incubations resulted in the modification of NDUFS1 subunit in complex I. Glutathionylation could be reversed by reducing agents, restoring the deglutathionylated state of NDUFS1 and the activity of the complex. Reduction of glutathionyl moieties in complex I also significantly decreased ROS production by RET from GPD and PRODH. Overall, these findings demonstrate that the modification of NDUFS1 can result in increased ROS production during RET from the UQ pool, which has implications for understanding the relationship between mitochondrial glutathionylation reactions and induction of oxidative distress in several pathologies.
Collapse
Affiliation(s)
| | | | | | - Ryan J. Mailloux
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, Montreal, QC H9X 3V9, Canada
| |
Collapse
|
8
|
Leduc-Gaudet JP, Hussain SN, Gouspillou G. Parkin: A potential target to promote healthy aging. J Physiol 2022; 600:3405-3421. [PMID: 35691026 DOI: 10.1113/jp282567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/08/2022] [Indexed: 11/08/2022] Open
Abstract
Parkin is an E3 ubiquitin ligase mostly known for its role in regulating the removal of defective mitochondria via mitophagy. However, increasing experimental evidence that Parkin regulates several other aspects of mitochondrial biology in addition to its role in mitophagy has emerged over the past two decades. Indeed, Parkin has been shown to regulate mitochondrial biogenesis and dynamics and mitochondrial-derived vesicle formation, suggesting that Parkin plays key roles in maintaining healthy mitochondria. While Parkin is commonly described as a cytosolic E3 ubiquitin ligase, Parkin was also detected in other cellular compartments, including the nucleus, where it regulates transcription factors and acts as a transcription factor itself. New evidence also suggests that Parkin overexpression can be leveraged to delay aging. In D. melanogaster, for example, Parkin overexpression extends lifespan. In mammals, Parkin overexpression delays hallmarks of aging in several tissues and cell types. Parkin overexpression also confers protection in various models of cellular senescence and neurological disorders closely associated with aging, such as Alzheimer's and Parkinson's diseases. Recently, Parkin overexpression has also been shown to suppress tumor growth. In this review, we discuss newly emerging biological roles of Parkin as a modulator of cellular homeostasis, survival, and healthy aging, and we explore potential mechanisms through which Parkin exerts its beneficial effects on cellular health. Abstract figure legend Parkin: A potential target to promote healthy aging Illustration of key aspects of Parkin biology, including Parkin function and cellular localization and key roles in the regulation of mitochondrial quality control. The organs and systems in which Parkin overexpression was shown to exert protective effects relevant to the promotion of healthy aging are highlighted in the black rectangle at the bottom of the Figure. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jean-Philippe Leduc-Gaudet
- Department of Biomedical Sciences, Veneto Institute of Molecular Medicine, University of Padova, Padova, Italy.,Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC, Canada.,Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada.,Département des sciences de l'activité physique, Faculté des sciences, Université du Québec à Montréal (UQAM), Montréal, QC, Canada
| | - Sabah Na Hussain
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC, Canada.,Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Gilles Gouspillou
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC, Canada.,Département des sciences de l'activité physique, Faculté des sciences, Université du Québec à Montréal (UQAM), Montréal, QC, Canada
| |
Collapse
|
9
|
Yadav P, Sharma P, Sundaram S, Venkatraman G, Bera AK, Karunagaran D. SLC7A11/ xCT is a target of miR-5096 and its restoration partially rescues miR-5096-mediated ferroptosis and anti-tumor effects in human breast cancer cells. Cancer Lett 2021; 522:211-224. [PMID: 34571083 DOI: 10.1016/j.canlet.2021.09.033] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Revised: 09/08/2021] [Accepted: 09/21/2021] [Indexed: 01/15/2023]
Abstract
Breast cancer cells evade cell death by overexpressing SLC7A11, which functions by transporting cystine into cells in exchange for intracellular glutamate facilitating glutathione synthesis and reducing reactive oxygen species (ROS)-mediated stress. Using an in silico approach, we predicted an miRNA (miR-5096) that can target and downregulate SLC7A11. We demonstrated SLC7A11 as a target of miR-5096 by 3'UTR luciferase assay and further validated it by identifying reduced mRNA and protein levels of SLC7A11 upon miR-5096 overexpression. miR-5096-induced ferroptotic cell death in human breast cancer cells was confirmed by concurrently increased ROS, OH-, lipid ROS, and iron accumulation levels and decreased GSH and mitochondrial membrane potential (MitoTracker™ Orange) with mitochondrial shrinkage and partial cristae loss (observed by TEM). miR-5096 inhibited colony formation, transwell migration, and breast cancer cell invasion, whereas antimiR-5096 promoted these tumorigenic properties. Ectopic expression of SLC7A11 partly reversed miR-5096-mediated effects on cell survival, ROS, lipid peroxides, iron accumulation, GSH, hydroxyl radicals, mitochondrial membrane potential, and colony formation. miR-5096 modulated the expression of epithelial-mesenchymal transition markers in vitro and inhibited the metastatic potential of MDA-MB-231 cells in a tumor xenograft model of zebrafish larvae. Our results demonstrate that miR-5096 is a tumor-suppressive miRNA in breast cancer cells, and this paper discusses its therapeutic implications.
Collapse
Affiliation(s)
- Poonam Yadav
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Priyanshu Sharma
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Sandhya Sundaram
- Department of Pathology, Sri Ramachandra Medical College and Research Institute, Sri Ramachandra University, Porur, Chennai, 600116, India
| | - Ganesh Venkatraman
- Department of Human Genetics, Faculty of Biomedical Sciences, Technology and Research, Sri Ramachandra Institute of Higher Education and Research, Chennai, 600116, India
| | - Amal Kanti Bera
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Devarajan Karunagaran
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| |
Collapse
|
10
|
Shi Y, He R, Yang Y, He Y, Zhan L, Wei B. Potential relationship between Sirt3 and autophagy in ovarian cancer. Oncol Lett 2020; 20:162. [PMID: 32934730 PMCID: PMC7471650 DOI: 10.3892/ol.2020.12023] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023] Open
Abstract
Sirtuin 3 (Sirt3) is an important member of the sirtuin protein family. It is a deacetylase that was previously reported to modulate the level of reactive oxygen species (ROS) production and limit the extent of oxidative damage in cellular components. As an important member of the class III type of histone deacetylases, Sirt3 has also been documented to mediate nuclear gene expression, metabolic control, neuroprotection, cell cycle and proliferation. In ovarian cancer (OC), Sirt3 has been reported to regulate cellular metabolism, apoptosis and autophagy. Sirt3 can regulate autophagy through a variety of different molecular signaling pathways, including the p62, 5'AMP-activated protein kinase and mitochondrial ROS-superoxide dismutase pathways. However, autophagy downstream of Sirt3 and its association with OC remains poorly understood. In the present review, the known characteristics of Sirt3 and autophagy were outlined, and their potential functional roles were discussed. Following a comprehensive analysis of the current literature, Sirt3 and autophagy may either serve positive or negative roles in the regulation of OC. Therefore, it is important to identify the appropriate expression level of Sirt3 to control the activation of autophagy in OC cells. This strategy may prove to be a novel therapeutic method to reduce the mortality of patients with OC. Finally, potential research directions into the association between Sirt3 and other signaling pathways were provided.
Collapse
Affiliation(s)
- Yuchuan Shi
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Runhua He
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Yu Yang
- Department of Cardiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Yu He
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| | - Lei Zhan
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China.,Department of Obstetrics and Gynecology, Reproductive Medicine Center, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230022, P.R. China
| | - Bing Wei
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, P.R. China
| |
Collapse
|
11
|
Daniel T, Faruq HM, Laura Magdalena J, Manuela G, Christopher Horst L. Role of GSH and Iron-Sulfur Glutaredoxins in Iron Metabolism-Review. Molecules 2020; 25:E3860. [PMID: 32854270 PMCID: PMC7503856 DOI: 10.3390/molecules25173860] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/19/2020] [Accepted: 08/22/2020] [Indexed: 12/26/2022] Open
Abstract
Glutathione (GSH) was initially identified and characterized for its redox properties and later for its contributions to detoxification reactions. Over the past decade, however, the essential contributions of glutathione to cellular iron metabolism have come more and more into focus. GSH is indispensable in mitochondrial iron-sulfur (FeS) cluster biosynthesis, primarily by co-ligating FeS clusters as a cofactor of the CGFS-type (class II) glutaredoxins (Grxs). GSH is required for the export of the yet to be defined FeS precursor from the mitochondria to the cytosol. In the cytosol, it is an essential cofactor, again of the multi-domain CGFS-type Grxs, master players in cellular iron and FeS trafficking. In this review, we summarize the recent advances and progress in this field. The most urgent open questions are discussed, such as the role of GSH in the export of FeS precursors from mitochondria, the physiological roles of the CGFS-type Grx interactions with BolA-like proteins and the cluster transfer between Grxs and recipient proteins.
Collapse
Affiliation(s)
- Trnka Daniel
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, University of Greifswald, 17475 Greifswald, Germany; (T.D.); (H.M.F.); (J.L.M.); (G.M.)
| | - Hossain Md Faruq
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, University of Greifswald, 17475 Greifswald, Germany; (T.D.); (H.M.F.); (J.L.M.); (G.M.)
| | - Jordt Laura Magdalena
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, University of Greifswald, 17475 Greifswald, Germany; (T.D.); (H.M.F.); (J.L.M.); (G.M.)
| | - Gellert Manuela
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, University of Greifswald, 17475 Greifswald, Germany; (T.D.); (H.M.F.); (J.L.M.); (G.M.)
| | - Lillig Christopher Horst
- Christopher Horst Lillig, Institute for Medical Biochemistry and Molecular Biology, University Medicine Greifswald, Ferdinand-Sauerbruch-Straße, 17475 Greifswald, Germany
| |
Collapse
|
12
|
Sun Y, Deng R, Zhang C. Erastin induces apoptotic and ferroptotic cell death by inducing ROS accumulation by causing mitochondrial dysfunction in gastric cancer cell HGC‑27. Mol Med Rep 2020; 22:2826-2832. [PMID: 32945484 PMCID: PMC7453531 DOI: 10.3892/mmr.2020.11376] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 05/27/2020] [Indexed: 12/16/2022] Open
Abstract
Erastin, a classical inducer of non-apoptotic cell death, exerts cytotoxicity in several types of cancer cells, including gastric cancer cells, by depleting glutathione, which is a primary cellular antioxidant, thus causing reactive oxygen species (ROS) accumulation. Although numerous studies have focused on the non-apoptotic cell death induced by erastin, whether erastin induces apoptosis remains unknown. The present study confirmed the cytotoxicity of erastin in HGC-27 cells and used a 30% inhibitory concentration (IC30, approximately 6.23 µM) for further analysis. The cell cycle analysis revealed that 6.23 µM of erastin inhibited proliferation by blocking the cell cycle at the G1/G0 phase. Further analysis also showed that 6.23 µM of erastin clearly inhibited HGC-27 malignant behaviors, including migration, invasion, colony formation and tumor formation in soft agar. The observation of ROS accumulation due to erastin treatment led to determination of the effects of erastin on mitochondrial function and, as expected, erastin treatment decreased transcriptional activity and ATP production in mitochondria and disrupted the mitochondrial potential; these effects were reversed by the addition of the ROS scavenger NAC. To evaluate the effect of erastin in inducing apoptosis, HGC-27 cells were treated with 6.23 µM of erastin for 7 days and then analyzed. Evident apoptotic cell death was induced by erastin and this apoptosis was reversed by the addition of an apoptosis inhibitor (zVAD) or NAC but not by the addition of a ferroptosis inhibitor (ferrostatin-1). Furthermore, the detection of caspase-3 and poly (adenosine diphosphate-ribose) polymerase (PARP) also confirmed that treatment with erastin promoted the cleavage of caspase-3 and PARP, which are hallmarks of apoptosis. Taken together, the present study revealed that a low dose of erastin inhibited malignant behavior and induced apoptosis by causing mitochondrial dysfunction.
Collapse
Affiliation(s)
- Yingwei Sun
- Department of Pharmacy, The Teaching Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Rongrong Deng
- Medicine School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 610072, P.R. China
| | - Cuiwei Zhang
- Department of Pathology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
13
|
Molecular basis for the distinct functions of redox-active and FeS-transfering glutaredoxins. Nat Commun 2020; 11:3445. [PMID: 32651396 PMCID: PMC7351949 DOI: 10.1038/s41467-020-17323-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Accepted: 06/18/2020] [Indexed: 02/08/2023] Open
Abstract
Despite their very close structural similarity, CxxC/S-type (class I) glutaredoxins (Grxs) act as oxidoreductases, while CGFS-type (class II) Grxs act as FeS cluster transferases. Here we show that the key determinant of Grx function is a distinct loop structure adjacent to the active site. Engineering of a CxxC/S-type Grx with a CGFS-type loop switched its function from oxidoreductase to FeS transferase. Engineering of a CGFS-type Grx with a CxxC/S-type loop abolished FeS transferase activity and activated the oxidative half reaction of the oxidoreductase. The reductive half-reaction, requiring the interaction with a second GSH molecule, was enabled by switching additional residues in the active site. We explain how subtle structural differences, mostly depending on the structure of one particular loop, act in concert to determine Grx function.
Collapse
|
14
|
Matsui R, Ferran B, Oh A, Croteau D, Shao D, Han J, Pimentel DR, Bachschmid MM. Redox Regulation via Glutaredoxin-1 and Protein S-Glutathionylation. Antioxid Redox Signal 2020; 32:677-700. [PMID: 31813265 PMCID: PMC7047114 DOI: 10.1089/ars.2019.7963] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Significance: Over the past several years, oxidative post-translational modifications of protein cysteines have been recognized for their critical roles in physiology and pathophysiology. Cells have harnessed thiol modifications involving both oxidative and reductive steps for signaling and protein processing. One of these stages requires oxidation of cysteine to sulfenic acid, followed by two reduction reactions. First, glutathione (reduced glutathione [GSH]) forms a S-glutathionylated protein, and second, enzymatic or chemical reduction removes the modification. Under physiological conditions, these steps confer redox signaling and protect cysteines from irreversible oxidation. However, oxidative stress can overwhelm protein S-glutathionylation and irreversibly modify cysteine residues, disrupting redox signaling. Critical Issues: Glutaredoxins mainly catalyze the removal of protein-bound GSH and help maintain protein thiols in a highly reduced state without exerting direct antioxidant properties. Conversely, glutathione S-transferase (GST), peroxiredoxins, and occasionally glutaredoxins can also catalyze protein S-glutathionylation, thus promoting a dynamic redox environment. Recent Advances: The latest studies of glutaredoxin-1 (Glrx) transgenic or knockout mice demonstrate important distinct roles of Glrx in a variety of pathologies. Endogenous Glrx is essential to maintain normal hepatic lipid homeostasis and prevent fatty liver disease. Further, in vivo deletion of Glrx protects lungs from inflammation and bacterial pneumonia-induced damage, attenuates angiotensin II-induced cardiovascular hypertrophy, and improves ischemic limb vascularization. Meanwhile, exogenous Glrx administration can reverse pathological lung fibrosis. Future Directions: Although S-glutathionylation modifies many proteins, these studies suggest that S-glutathionylation and Glrx regulate specific pathways in vivo, and they implicate Glrx as a potential novel therapeutic target to treat diverse disease conditions. Antioxid. Redox Signal. 32, 677-700.
Collapse
Affiliation(s)
- Reiko Matsui
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Beatriz Ferran
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Albin Oh
- Cardiology, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Dominique Croteau
- Cardiology, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Di Shao
- Helens Clinical Research Center, Chongqing, China
| | - Jingyan Han
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - David Richard Pimentel
- Cardiology, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Markus Michael Bachschmid
- Vascular Biology Section, Department of Medicine, Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| |
Collapse
|
15
|
Lyu C, Webber DM, MacLeod SL, Hobbs CA, Li M. Gene-by-gene interactions associated with the risk of conotruncal heart defects. Mol Genet Genomic Med 2020; 8:e1010. [PMID: 31851787 PMCID: PMC6978401 DOI: 10.1002/mgg3.1010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/11/2019] [Accepted: 09/25/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The development of conotruncal heart defects (CTDs) involves a complex relationship among genetic variants and maternal lifestyle factors. In this article, we focused on the interactions between 13 candidate genes within folate, homocysteine, and transsulfuration pathways for potential association with CTD risk. METHODS Targeted sequencing was used for 328 case-parental triads enrolled in the National Birth Defects Prevention Study (NBDPS). To evaluate the interaction of two genes, we applied a conditional logistic regression model for all possible SNP pairs within two respective genes by contrasting the affected infants with their pseudo-controls. The findings were replicated in an independent sample of 86 NBDPS case-parental triads genotyped by DNA microarrays. The results of two studies were further integrated by a fixed-effect meta-analysis. RESULTS One SNP pair (i.e., rs4764267 and rs6556883) located in gene MGST1 and GLRX, respectively, was found to be associated with CTD risk after multiple testing adjustment using simpleM, a modified Bonferroni correction approach (nominal p-value of 4.62e-06; adjusted p-value of .04). Another SNP pair (i.e., rs11892646 and rs56219526) located in gene DNMT3A and MTRR, respectively, achieved marginal significance after multiple testing adjustment (adjusted p-value of .06). CONCLUSION Further studies with larger sample sizes are needed to confirm and elucidate these potential interactions.
Collapse
Affiliation(s)
- Chen Lyu
- Department of Epidemiology and BiostatisticsIndiana UniversityBloomingtonINUSA
| | - Daniel M. Webber
- Department of Pathology & ImmunologyWashington University at St LouisSaint LouisMOUSA
| | | | | | - Ming Li
- Department of Epidemiology and BiostatisticsIndiana UniversityBloomingtonINUSA
| | | |
Collapse
|
16
|
Control of doxorubicin-induced, reactive oxygen-related apoptosis by glutathione peroxidase 1 in cardiac fibroblasts. Biochem Biophys Rep 2019; 21:100709. [PMID: 31799454 PMCID: PMC6881695 DOI: 10.1016/j.bbrep.2019.100709] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 11/19/2019] [Indexed: 12/16/2022] Open
Abstract
Reactive oxygen formation plays a mechanistic role in the cardiotoxicity of doxorubicin, a chemotherapeutic agent that remains an important component of treatment programs for breast cancer and hematopoietic malignancies. To examine the role of doxorubicin-induced reactive oxygen species (ROS) in drug-related cardiac apoptosis, murine embryonic fibroblast cell lines were derived from the hearts of glutathione peroxidase 1 (Gpx-1) knockout mice. Cells from homozygous Gpx-1 knockout mice and parental animals were propagated with (Se+) and without (Se-) 100 nM sodium selenite. Activity levels of the peroxide detoxifying selenoprotein glutathione peroxidase (GSHPx) were marginally detectable (<1.6 nmol/min/mg) in fibroblasts from homozygous knockout animals whether or not cells were supplemented with selenium. GSHPx activity in Se- cells from parental murine fibroblasts was also <1.6 nmol/min/mg, whereas GSHPx levels in Se+ parental murine fibroblasts were 12.9 ± 2.7 nmol/min/mg (mean ± SE; P < 0.05). Catalase, superoxide dismutase, glutathione reductase, glutathione S-transferase, glucose 6-phosphate dehydrogenase, and reduced glutathione activities did not differ amongst the four cell lines. Reactive oxygen production increased from 908 ± 122 (arbitrary units) for untreated control cells to 1668 ± 54 following exposure to 1 μM doxorubicin for 24 h in parental fibroblasts not supplemented with selenium (P < 0.03); reactive oxygen formation in doxorubicin-treated parental fibroblasts propagated in selenium was 996 ± 69 (P = not significant compared to untreated control cells). Reactive oxygen levels in homozygous Gpx-1 knockout fibroblasts, irrespective of selenium supplementation status, were increased and equivalent to that in selenium deficient wild type fibroblasts. When cardiac fibroblasts were exposed to doxorubicin (0.05 μM) for 96 h and examined for cell cycle alterations by flow cytometry, and apoptosis by TUNEL assay, marked G2 arrest and TUNEL positivity were observed in knockout fibroblasts in the presence or absence of supplemental selenium, and in parental fibroblasts propagated without selenium. Parental fibroblasts propagated with selenium and exposed to the same concentration of doxorubicin demonstrated modest TUNEL positivity and substantially diminished amounts of low molecular weight DNA. These results were replicated in cardiac fibroblasts exposed to doxorubicin (1–2 μM) for 2 h (to mimic clinical drug dosing schedules) and examined 96 h following initiation of drug exposure. Doxorubicin uptake in cardiac fibroblasts was similar irrespective of the mRNA expression level or activity of GSHPx. These experiments suggest that the intracellular levels of doxorubicin-induced reactive oxygen species (ROS) are modulated by GSHPx and play an important role in doxorubicin-related apoptosis and altered cell cycle progression in murine cardiac fibroblasts.
Collapse
|
17
|
Sex-dependent Differences in the Bioenergetics of Liver and Muscle Mitochondria from Mice Containing a Deletion for glutaredoxin-2. Antioxidants (Basel) 2019; 8:antiox8080245. [PMID: 31357416 PMCID: PMC6720827 DOI: 10.3390/antiox8080245] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/18/2019] [Accepted: 07/24/2019] [Indexed: 02/07/2023] Open
Abstract
Our group recently published a study demonstrating that deleting the gene encoding the matrix thiol oxidoreductase, glutaredoxin-2 (GRX2), alters the bioenergetics of mitochondria isolated from male C57BL/6N mice. Here, we conducted a similar study, examining H2O2 production and respiration in mitochondria isolated from female mice heterozygous (GRX2+/−) or homozygous (GRX2−/−) for glutaredoxin-2. First, we observed that deleting the Grx2 gene does not alter the rate of H2O2 production in liver and muscle mitochondria oxidizing pyruvate, α-ketoglutarate, or succinate. Examination of the rates of H2O2 release from liver mitochondria isolated from male and female mice revealed that (1) sex has an impact on the rate of ROS production by liver and muscle mitochondria and (2) loss of GRX2 only altered ROS release in mitochondria collected from male mice. Assessment of the bioenergetics of these mitochondria revealed that loss of GRX2 increased proton leak-dependent and phosphorylating respiration in liver mitochondria isolated from female mice but did not alter rates of respiration in liver mitochondria from male mice. Furthermore, we found that deleting the Grx2 gene did not alter rates of respiration in muscle mitochondria collected from female mice. This contrasts with male mice where loss of GRX2 substantially augmented proton leaks and ADP-stimulated respiration. Our findings indicate that some fundamental sexual dimorphisms exist between GRX2-deficient male and female rodents.
Collapse
|
18
|
Mitochondrial dynamics in exercise physiology. Pflugers Arch 2019; 472:137-153. [DOI: 10.1007/s00424-019-02258-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2019] [Accepted: 01/17/2019] [Indexed: 12/11/2022]
|
19
|
Ferdinandy P, Baczkó I, Bencsik P, Giricz Z, Görbe A, Pacher P, Varga ZV, Varró A, Schulz R. Definition of hidden drug cardiotoxicity: paradigm change in cardiac safety testing and its clinical implications. Eur Heart J 2018; 40:1771-1777. [PMID: 29982507 PMCID: PMC6554653 DOI: 10.1093/eurheartj/ehy365] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 03/12/2018] [Accepted: 06/11/2018] [Indexed: 12/18/2022] Open
Abstract
Unexpected cardiac adverse effects are the leading causes of discontinuation of clinical trials and withdrawal of drugs from the market. Since the original observations in the mid-90s, it has been well established that cardiovascular risk factors and comorbidities (such as ageing, hyperlipidaemia, and diabetes) and their medications (e.g. nitrate tolerance, adenosine triphosphate-dependent potassium inhibitor antidiabetic drugs, statins, etc.) may interfere with cardiac ischaemic tolerance and endogenous cardioprotective signalling pathways. Indeed drugs may exert unwanted effects on the diseased and treated heart that is hidden in the healthy myocardium. Hidden cardiotoxic effects may be due to (i) drug-induced enhancement of deleterious signalling due to ischaemia/reperfusion injury and/or the presence of risk factors and/or (ii) inhibition of cardioprotective survival signalling pathways, both of which may lead to ischaemia-related cell death and/or pro-arrhythmic effects. This led to a novel concept of ‘hidden cardiotoxicity’, defined as cardiotoxity of a drug that manifests only in the diseased heart with e.g. ischaemia/reperfusion injury and/or in the presence of its major comorbidities. Little is known on the mechanism of hidden cardiotoxocity, moreover, hidden cardiotoxicity cannot be revealed by the routinely used non-clinical cardiac safety testing methods on healthy animals or tissues. Therefore, here, we emphasize the need for development of novel cardiac safety testing platform involving combined experimental models of cardiac diseases (especially myocardial ischaemia/reperfusion and ischaemic conditioning) in the presence and absence of major cardiovascular comorbidities and/or cotreatments.
Collapse
Affiliation(s)
- Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - István Baczkó
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, Szeged, Hungary
| | | | - Zoltán Giricz
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Pharmahungary Group, Hajnoczy u. 6, Szeged, Hungary
| | - Pál Pacher
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Fishers Lane, Bethesda, MD, USA
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Nagyvárad tér 4, Budapest, Hungary
- Laboratory of Cardiovascular Physiology and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health, Fishers Lane, Bethesda, MD, USA
| | - András Varró
- Department of Pharmacology and Pharmacotherapy, University of Szeged, Dóm tér 12, Szeged, Hungary
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University of Giessen, Aulweg 129, Giessen, Germany
| |
Collapse
|
20
|
Giorgi C, Marchi S, Simoes IC, Ren Z, Morciano G, Perrone M, Patalas-Krawczyk P, Borchard S, Jȩdrak P, Pierzynowska K, Szymański J, Wang DQ, Portincasa P, Wȩgrzyn G, Zischka H, Dobrzyn P, Bonora M, Duszynski J, Rimessi A, Karkucinska-Wieckowska A, Dobrzyn A, Szabadkai G, Zavan B, Oliveira PJ, Sardao VA, Pinton P, Wieckowski MR. Mitochondria and Reactive Oxygen Species in Aging and Age-Related Diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2018; 340:209-344. [PMID: 30072092 PMCID: PMC8127332 DOI: 10.1016/bs.ircmb.2018.05.006] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Aging has been linked to several degenerative processes that, through the accumulation of molecular and cellular damage, can progressively lead to cell dysfunction and organ failure. Human aging is linked with a higher risk for individuals to develop cancer, neurodegenerative, cardiovascular, and metabolic disorders. The understanding of the molecular basis of aging and associated diseases has been one major challenge of scientific research over the last decades. Mitochondria, the center of oxidative metabolism and principal site of reactive oxygen species (ROS) production, are crucial both in health and in pathogenesis of many diseases. Redox signaling is important for the modulation of cell functions and several studies indicate a dual role for ROS in cell physiology. In fact, high concentrations of ROS are pathogenic and can cause severe damage to cell and organelle membranes, DNA, and proteins. On the other hand, moderate amounts of ROS are essential for the maintenance of several biological processes, including gene expression. In this review, we provide an update regarding the key roles of ROS-mitochondria cross talk in different fundamental physiological or pathological situations accompanying aging and highlighting that mitochondrial ROS may be a decisive target in clinical practice.
Collapse
Affiliation(s)
- Carlotta Giorgi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Saverio Marchi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Ines C.M. Simoes
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Ziyu Ren
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
| | - Giampaolo Morciano
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
- Maria Pia Hospital, GVM Care & Research, Torino, Italy
| | - Mariasole Perrone
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | - Paulina Patalas-Krawczyk
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Sabine Borchard
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - Paulina Jȩdrak
- Department of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | | | - Jȩdrzej Szymański
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - David Q. Wang
- Department of Medicine, Division of Gastroenterology and Liver Diseases, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Dept. of Biomedical Sciences & Human Oncology, University of Bari "Aldo Moro" Medical School, Bari, Italy
| | - Grzegorz Wȩgrzyn
- Department of Molecular Biology, University of Gdańsk, Gdańsk, Poland
| | - Hans Zischka
- Institute of Molecular Toxicology and Pharmacology, Helmholtz Center Munich, German Research Center for Environmental Health, Neuherberg, Germany
- Institute of Toxicology and Environmental Hygiene, Technical University Munich, Munich, Germany
| | - Pawel Dobrzyn
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Massimo Bonora
- Departments of Cell Biology and Gottesman Institute for Stem Cell & Regenerative Medicine Research, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Jerzy Duszynski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| | - Alessandro Rimessi
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
| | | | | | - Gyorgy Szabadkai
- Department of Cell and Developmental Biology, Consortium for Mitochondrial Research, University College London, London, United Kingdom
- The Francis Crick Institute, London, United Kingdom
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Barbara Zavan
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
- Department of Biomedical Sciences, University of Padua, Padua, Italy
| | - Paulo J. Oliveira
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, Biocant Park, University of Coimbra, Cantanhede, Portugal
| | - Vilma A. Sardao
- CNC - Center for Neuroscience and Cell Biology, UC-Biotech, Biocant Park, University of Coimbra, Cantanhede, Portugal
| | - Paolo Pinton
- Department of Morphology Surgery and Experimental Medicine, Section of Pathology Oncology and Experimental Biology, Interdisciplinary Center for the Study of Inflammation (ICSI), Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Ferrara, Italy
- Cecilia Hospital, GVM Care & Research, 48033 Cotignola, Ravenna, Italy
| | - Mariusz R. Wieckowski
- Department of Biochemistry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
21
|
Chalker J, Gardiner D, Kuksal N, Mailloux RJ. Characterization of the impact of glutaredoxin-2 (GRX2) deficiency on superoxide/hydrogen peroxide release from cardiac and liver mitochondria. Redox Biol 2018; 15:216-227. [PMID: 29274570 PMCID: PMC5773472 DOI: 10.1016/j.redox.2017.12.006] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 12/07/2017] [Accepted: 12/13/2017] [Indexed: 01/30/2023] Open
Abstract
Mitochondria are critical sources of hydrogen peroxide (H2O2), an important secondary messenger in mammalian cells. Recent work has shown that O2•-/H2O2 emission from individual sites of production in mitochondria is regulated by protein S-glutathionylation. Here, we conducted the first examination of O2•-/H2O2 release rates from cardiac and liver mitochondria isolated from mice deficient for glutaredoxin-2 (GRX2), a matrix-associated thiol oxidoreductase that facilitates the S-glutathionylation and deglutathionylation of proteins. Liver mitochondria isolated from mice heterozygous (GRX2+/-) and homozygous (GRX2-/-) for glutaredoxin-2 displayed a significant decrease in O2•-/H2O2 release when oxidizing pyruvate or 2-oxoglutarate. The genetic deletion of the Grx2 gene was associated with increased protein expression of pyruvate dehydrogenase (PDH) but not 2-oxoglutarate dehydrogenase (OGDH). By contrast, O2•-/H2O2 production was augmented in cardiac mitochondria from GRX2+/- and GRX2-/- mice metabolizing pyruvate or 2-oxoglutarate which was associated with decreased PDH and OGDH protein levels. ROS production was augmented in liver and cardiac mitochondria metabolizing succinate. Inhibitor studies revealed that OGDH and Complex III served as high capacity ROS release sites in liver mitochondria. By contrast, Complex I and Complex III were found to be the chief O2•-/H2O2 emitters in cardiac mitochondria. These findings identify an essential role for GRX2 in regulating O2•-/H2O2 release from mitochondria in liver and cardiac tissue. Our results demonstrate that the GRX2-mediated regulation of O2•-/H2O2 release through the S-glutathionylation of mitochondrial proteins may play an integral role in controlling cellular ROS signaling.
Collapse
Affiliation(s)
- Julia Chalker
- Memorial University of Newfoundland, Department of Biochemistry, St. John's, Newfoundland, Canada
| | - Danielle Gardiner
- Memorial University of Newfoundland, Department of Biochemistry, St. John's, Newfoundland, Canada
| | - Nidhi Kuksal
- Memorial University of Newfoundland, Department of Biochemistry, St. John's, Newfoundland, Canada
| | - Ryan J Mailloux
- Memorial University of Newfoundland, Department of Biochemistry, St. John's, Newfoundland, Canada.
| |
Collapse
|
22
|
Wang Y, Lei T, Yuan J, Wu Y, Shen X, Gao J, Feng W, Lu Z. GCN2 deficiency ameliorates doxorubicin-induced cardiotoxicity by decreasing cardiomyocyte apoptosis and myocardial oxidative stress. Redox Biol 2018; 17:25-34. [PMID: 29660505 PMCID: PMC6006681 DOI: 10.1016/j.redox.2018.04.009] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 04/04/2018] [Accepted: 04/06/2018] [Indexed: 01/17/2023] Open
Abstract
The clinical use of doxorubicin for cancer therapy is limited by its cardiotoxicity, which involves cardiomyocyte apoptosis and oxidative stress. Previously, we showed that general control nonderepressible 2 (GCN2), an eukaryotic initiation factor 2α (eIF2α) kinase, impairs the ventricular adaptation to chronic pressure overload by affecting cardiomyocyte apoptosis. However, the impact of GCN2 on Dox-induced cardiotoxicity has not been investigated. In the present study, we treated wild type (WT) and Gcn2−/− mice with four intraperitoneal injections (5 mg/kg/week) to induce cardiomyopathy. After Dox treatment, Gcn2−/− mice developed less contractile dysfunction, myocardial fibrosis, apoptosis, and oxidative stress compared with WT mice. In the hearts of the Dox-treated mice, GCN2 deficiency attenuated eIF2α phosphorylation and induction of its downstream targets, activating transcription factor 4 (ATF4) and C/EBP homologous protein (CHOP), and preserved the expression of anti-apoptotic factor Bcl-2 and mitochondrial uncoupling protein-2(UCP2). Furthermore, we found that GCN2 knockdown attenuated, whereas GCN2 overexpression exacerbated, Dox-induced cell death, oxidative stress and reduction of Bcl-2 and UCP2 expression through the eIF2α-CHOP-dependent pathway in H9C2 cells. Collectively, our data provide solid evidence that GCN2 has a marked effect on Dox induced myocardial apoptosis and oxidative stress. Our findings suggest that strategies to inhibit GCN2 activity in cardiomyocyte may provide a novel approach to attenuate Dox-related cardiotoxicity. GCN2 deficiency ameliorates doxorubicin-induced cardiac dysfunction. GCN2 promotes doxorubicin-induced cardiomyocyte apoptosis and oxidative stress. GCN2 decreases Bcl-2 and UCP2 expression via a CHOP dependent manner. Knockdown of UCP2 exacerbated doxorubicin-induced cell death and oxidative stress.
Collapse
Affiliation(s)
- Yue Wang
- College of Life Science, University of Chinese Academy of Sciences, 19 A Yuquanlu, Beijing 100049, China
| | - Tong Lei
- College of Life Science, University of Chinese Academy of Sciences, 19 A Yuquanlu, Beijing 100049, China
| | - Juntao Yuan
- College of Life Science, University of Chinese Academy of Sciences, 19 A Yuquanlu, Beijing 100049, China
| | - Yongguang Wu
- College of Life Science, University of Chinese Academy of Sciences, 19 A Yuquanlu, Beijing 100049, China
| | - Xiyue Shen
- College of Life Science, University of Chinese Academy of Sciences, 19 A Yuquanlu, Beijing 100049, China
| | - Junling Gao
- College of Life Science, University of Chinese Academy of Sciences, 19 A Yuquanlu, Beijing 100049, China
| | - Wei Feng
- College of Life Science, University of Chinese Academy of Sciences, 19 A Yuquanlu, Beijing 100049, China
| | - Zhongbing Lu
- College of Life Science, University of Chinese Academy of Sciences, 19 A Yuquanlu, Beijing 100049, China.
| |
Collapse
|
23
|
Bothiraja C, Rajput N, Poudel I, Rajalakshmi S, Panda B, Pawar A. Development of novel biofunctionalized chitosan decorated nanocochleates as a cancer targeted drug delivery platform. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:447-461. [DOI: 10.1080/21691401.2018.1430584] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- C. Bothiraja
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Pune, India
| | - Neeti Rajput
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Pune, India
| | - Ishwor Poudel
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Pune, India
| | - S. Rajalakshmi
- Department of Pharmaceutics, Dr D. Y. Patil College of Pharmacy, Pune, India
| | - Bijoy Panda
- Department of Clinical Pharmacy, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Pune, India
| | - Atmaram Pawar
- Department of Pharmaceutics, Poona College of Pharmacy, Bharati Vidyapeeth Deemed University, Pune, India
| |
Collapse
|
24
|
Kanaan GN, Ichim B, Gharibeh L, Maharsy W, Patten DA, Xuan JY, Reunov A, Marshall P, Veinot J, Menzies K, Nemer M, Harper ME. Glutaredoxin-2 controls cardiac mitochondrial dynamics and energetics in mice, and protects against human cardiac pathologies. Redox Biol 2017; 14:509-521. [PMID: 29101900 PMCID: PMC5675898 DOI: 10.1016/j.redox.2017.10.019] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 10/25/2017] [Indexed: 01/19/2023] Open
Abstract
Glutaredoxin 2 (GRX2), a mitochondrial glutathione-dependent oxidoreductase, is central to glutathione homeostasis and mitochondrial redox, which is crucial in highly metabolic tissues like the heart. Previous research showed that absence of Grx2, leads to impaired mitochondrial complex I function, hypertension and cardiac hypertrophy in mice but the impact on mitochondrial structure and function in intact cardiomyocytes and in humans has not been explored. We hypothesized that Grx2 controls cardiac mitochondrial dynamics and function in cellular and mouse models, and that low expression is associated with human cardiac dysfunction. Here we show that Grx2 absence impairs mitochondrial fusion, ultrastructure and energetics in primary cardiomyocytes and cardiac tissue. Moreover, provision of the glutathione precursor, N-acetylcysteine (NAC) to Grx2-/- mice did not restore glutathione redox or prevent impairments. Using genetic and histopathological data from the human Genotype-Tissue Expression consortium we demonstrate that low GRX2 is associated with fibrosis, hypertrophy, and infarct in the left ventricle. Altogether, GRX2 is important in the control of cardiac mitochondrial structure and function, and protects against human cardiac pathologies.
Collapse
Affiliation(s)
- Georges N Kanaan
- Department of Biochemistry, Microbiology and Immunology, and Ottawa Institute of Systems Biology, Faculty of Medicine, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | - Bianca Ichim
- Department of Biochemistry, Microbiology and Immunology, and Ottawa Institute of Systems Biology, Faculty of Medicine, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | - Lara Gharibeh
- Department of Biochemistry, Microbiology and Immunology, and Ottawa Institute of Systems Biology, Faculty of Medicine, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | - Wael Maharsy
- Department of Biochemistry, Microbiology and Immunology, and Ottawa Institute of Systems Biology, Faculty of Medicine, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | - David A Patten
- Department of Biochemistry, Microbiology and Immunology, and Ottawa Institute of Systems Biology, Faculty of Medicine, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | - Jian Ying Xuan
- Department of Biochemistry, Microbiology and Immunology, and Ottawa Institute of Systems Biology, Faculty of Medicine, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | - Arkadiy Reunov
- Ottawa Heart Institute, University of Ottawa, 40 Ruskin Street, Ottawa, ON, Canada K1Y 4W7
| | - Philip Marshall
- Interdisciplinary School of Health Sciences, University of Ottawa, Faculty of Health Sciences, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | - John Veinot
- Ottawa Heart Institute, University of Ottawa, 40 Ruskin Street, Ottawa, ON, Canada K1Y 4W7; The Ottawa Hospital, 501 Smyth Road, Ottawa, ON, Canada K1H8L6; Department of Pathology and Laboratory Medicine, and University of Ottawa, Faculty of Medicine, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | - Keir Menzies
- Department of Biochemistry, Microbiology and Immunology, and Ottawa Institute of Systems Biology, Faculty of Medicine, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5; Interdisciplinary School of Health Sciences, University of Ottawa, Faculty of Health Sciences, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | - Mona Nemer
- Department of Biochemistry, Microbiology and Immunology, and Ottawa Institute of Systems Biology, Faculty of Medicine, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, and Ottawa Institute of Systems Biology, Faculty of Medicine, 451 Smyth Road, Ottawa, ON, Canada K1H 8M5.
| |
Collapse
|
25
|
Shao D, Han J, Hou X, Fry J, Behring JB, Seta F, Long MT, Roy HK, Cohen RA, Matsui R, Bachschmid MM. Glutaredoxin-1 Deficiency Causes Fatty Liver and Dyslipidemia by Inhibiting Sirtuin-1. Antioxid Redox Signal 2017; 27:313-327. [PMID: 27958883 PMCID: PMC5563925 DOI: 10.1089/ars.2016.6716] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS Nonalcoholic fatty liver (NAFL) is a common liver disease associated with metabolic syndrome, obesity, and diabetes that is rising in prevalence worldwide. Various molecular perturbations of key regulators and enzymes in hepatic lipid metabolism cause NAFL. However, redox regulation through glutathione (GSH) adducts in NAFL remains largely elusive. Glutaredoxin-1 (Glrx) is a small thioltransferase that removes protein GSH adducts without having direct antioxidant properties. The liver contains abundant Glrx but its metabolic function is unknown. RESULTS Here we report that normal diet-fed Glrx-deficient mice (Glrx-/-) spontaneously develop obesity, hyperlipidemia, and hepatic steatosis by 8 months of age. Adenoviral Glrx repletion in the liver of Glrx-/- mice corrected lipid metabolism. Glrx-/- mice exhibited decreased sirtuin-1 (SirT1) activity that leads to hyperacetylation and activation of SREBP-1 and upregulation of key hepatic enzymes involved in lipid synthesis. We found that GSH adducts inhibited SirT1 activity in Glrx-/- mice. Hepatic expression of nonoxidizable cysteine mutant SirT1 corrected hepatic lipids in Glrx-/- mice. Wild-type mice fed high-fat diet develop metabolic syndrome, diabetes, and NAFL within several months. Glrx deficiency accelerated high-fat-induced NAFL and progression to steatohepatitis, manifested by hepatic damage and inflammation. INNOVATION These data suggest an essential role of hepatic Glrx in regulating SirT1, which controls protein glutathione adducts in the pathogenesis of hepatic steatosis. CONCLUSION We provide a novel redox-dependent mechanism for regulation of hepatic lipid metabolism, and propose that upregulation of hepatic Glrx may be a beneficial strategy for NAFL. Antioxid. Redox Signal. 27, 313-327.
Collapse
Affiliation(s)
- Di Shao
- 1 Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| | - Jingyan Han
- 1 Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| | - Xiuyun Hou
- 1 Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| | - Jessica Fry
- 1 Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| | - Jessica B Behring
- 1 Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| | - Francesca Seta
- 1 Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| | - Michelle T Long
- 3 Division of Gastroenterology, Boston Medical Center , Boston, Massachusetts
| | - Hemant K Roy
- 3 Division of Gastroenterology, Boston Medical Center , Boston, Massachusetts
| | - Richard A Cohen
- 1 Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts.,2 Cardiovascular Proteomics Center, Boston University School of Medicine , Boston, Massachusetts
| | - Reiko Matsui
- 1 Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts
| | - Markus M Bachschmid
- 1 Vascular Biology Section, Whitaker Cardiovascular Institute, Boston University School of Medicine , Boston, Massachusetts.,2 Cardiovascular Proteomics Center, Boston University School of Medicine , Boston, Massachusetts
| |
Collapse
|
26
|
O'Brien M, Chalker J, Slade L, Gardiner D, Mailloux RJ. Protein S-glutathionylation alters superoxide/hydrogen peroxide emission from pyruvate dehydrogenase complex. Free Radic Biol Med 2017; 106:302-314. [PMID: 28242228 DOI: 10.1016/j.freeradbiomed.2017.02.046] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Revised: 02/15/2017] [Accepted: 02/23/2017] [Indexed: 11/25/2022]
Abstract
Pyruvate dehydrogenase (Pdh) is a vital source of reactive oxygen species (ROS) in several different tissues. Pdh has also been suggested to serve as a mitochondrial redox sensor. Here, we report that O2•-/ H2O2 emission from pyruvate dehydrogenase (Pdh) is altered by S-glutathionylation. Glutathione disulfide (GSSG) amplified O2•-/ H2O2 production by purified Pdh during reverse electron transfer (RET) from NADH. Thiol oxidoreductase glutaredoxin-2 (Grx2) reversed these effects confirming that Pdh is a target for S-glutathionylation. S-glutathionylation had the opposite effect during forward electron transfer (FET) from pyruvate to NAD+ lowering O2•-/ H2O2 production. Immunoblotting for protein glutathione mixed disulfides (PSSG) following diamide treatment confirmed that purified Pdh can be S-glutathionylated. Similar observations were made with mouse liver mitochondria. S-glutathionylation catalysts diamide and disulfiram significantly reduced pyruvate or 2-oxoglutarate driven O2•-/ H2O2 production in liver mitochondria, results that were confirmed using various Pdh, 2-oxoglutarate dehydrogenase (Ogdh), and respiratory chain inhibitors. Immunoprecipitation of Pdh and Ogdh confirmed that either protein can be S-glutathionylated by diamide and disulfiram. Collectively, our results demonstrate that the S -glutathionylation of Pdh alters the amount of ROS formed by the enzyme complex. We also confirmed that Ogdh is controlled in a similar manner. Taken together, our results indicate that the redox sensing and ROS forming properties of Pdh and Ogdh are linked to S-glutathionylation.
Collapse
Affiliation(s)
- Marisa O'Brien
- Department of Biochemistry, Memorial University of Newfoundland, 230 Elizabeth Ave, St. John's, Newfoundland, Canada A1B 3X9
| | - Julia Chalker
- Department of Biochemistry, Memorial University of Newfoundland, 230 Elizabeth Ave, St. John's, Newfoundland, Canada A1B 3X9
| | - Liam Slade
- Department of Biochemistry, Memorial University of Newfoundland, 230 Elizabeth Ave, St. John's, Newfoundland, Canada A1B 3X9
| | - Danielle Gardiner
- Department of Biochemistry, Memorial University of Newfoundland, 230 Elizabeth Ave, St. John's, Newfoundland, Canada A1B 3X9
| | - Ryan J Mailloux
- Department of Biochemistry, Memorial University of Newfoundland, 230 Elizabeth Ave, St. John's, Newfoundland, Canada A1B 3X9.
| |
Collapse
|
27
|
Short JD, Downs K, Tavakoli S, Asmis R. Protein Thiol Redox Signaling in Monocytes and Macrophages. Antioxid Redox Signal 2016; 25:816-835. [PMID: 27288099 PMCID: PMC5107717 DOI: 10.1089/ars.2016.6697] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
SIGNIFICANCE Monocyte and macrophage dysfunction plays a critical role in a wide range of inflammatory disease processes, including obesity, impaired wound healing diabetic complications, and atherosclerosis. Emerging evidence suggests that the earliest events in monocyte or macrophage dysregulation include elevated reactive oxygen species production, thiol modifications, and disruption of redox-sensitive signaling pathways. This review focuses on the current state of research in thiol redox signaling in monocytes and macrophages, including (i) the molecular mechanisms by which reversible protein-S-glutathionylation occurs, (ii) the identification of bona fide S-glutathionylated proteins that occur under physiological conditions, and (iii) how disruptions of thiol redox signaling affect monocyte and macrophage functions and contribute to atherosclerosis. Recent Advances: Recent advances in redox biochemistry and biology as well as redox proteomic techniques have led to the identification of many new thiol redox-regulated proteins and pathways. In addition, major advances have been made in expanding the list of S-glutathionylated proteins and assessing the role that protein-S-glutathionylation and S-glutathionylation-regulating enzymes play in monocyte and macrophage functions, including monocyte transmigration, macrophage polarization, foam cell formation, and macrophage cell death. CRITICAL ISSUES Protein-S-glutathionylation/deglutathionylation in monocytes and macrophages has emerged as a new and important signaling paradigm, which provides a molecular basis for the well-established relationship between metabolic disorders, oxidative stress, and cardiovascular diseases. FUTURE DIRECTIONS The identification of specific S-glutathionylated proteins as well as the mechanisms that control this post-translational protein modification in monocytes and macrophages will facilitate the development of new preventive and therapeutic strategies to combat atherosclerosis and other metabolic diseases. Antioxid. Redox Signal. 25, 816-835.
Collapse
Affiliation(s)
- John D Short
- 1 Department of Pharmacology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Kevin Downs
- 2 Department of Cellular and Structural Biology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Sina Tavakoli
- 3 Department of Radiology, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| | - Reto Asmis
- 4 Department of Clinical Laboratory Sciences, University of Texas Health Science Center at San Antonio , San Antonio, Texas.,5 Department of Biochemistry, University of Texas Health Science Center at San Antonio , San Antonio, Texas
| |
Collapse
|
28
|
Cadete VJJ, Deschênes S, Cuillerier A, Brisebois F, Sugiura A, Vincent A, Turnbull D, Picard M, McBride HM, Burelle Y. Formation of mitochondrial-derived vesicles is an active and physiologically relevant mitochondrial quality control process in the cardiac system. J Physiol 2016; 594:5343-62. [PMID: 27311616 DOI: 10.1113/jp272703] [Citation(s) in RCA: 99] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 06/13/2016] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Mitochondrial-derived vesicle (MDV) formation occurs under baseline conditions and is rapidly upregulated in response to stress-inducing conditions in H9c2 cardiac myoblasts. In mice formation of MDVs occurs readily in the heart under normal healthy conditions while mitophagy is comparatively less prevalent. In response to acute stress induced by doxorubicin, mitochondrial dysfunction develops in the heart, triggering MDV formation and mitophagy. MDV formation is thus active in the cardiac system, where it probably constitutes a baseline housekeeping mechanism and a first line of defence against stress. ABSTRACT The formation of mitochondrial-derived vesicles (MDVs), a process inherited from bacteria, has emerged as a potentially important mitochondrial quality control (QC) mechanism to selectively deliver damaged material to lysosomes for degradation. However, the existence of this mechanism in various cell types, and its physiological relevance, remains unknown. Our aim was to investigate the dynamics of MDV formation in the cardiac system in vitro and in vivo. Immunofluorescence in cell culture, quantitative transmission electron microscopy and electron tomography in vivo were used to study MDV production in the cardiac system. We show that in cardiac cells MDV production occurs at baseline, is commensurate with the dependence of cells on oxidative metabolism, is more frequent than mitophagy and is up-regulated on the time scale of minutes to hours in response to prototypical mitochondrial stressors (antimycin-A, xanthine/xanthine oxidase). We further show that MDV production is up-regulated together with mitophagy in response to doxorubicin-induced mitochondrial and cardiac dysfunction. Here we provide the first quantitative data demonstrating that MDV formation is a mitochondrial QC operating in the heart.
Collapse
Affiliation(s)
| | - Sonia Deschênes
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada
| | | | | | - Ayumu Sugiura
- Neuromuscular Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Amy Vincent
- Welcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Doug Turnbull
- Welcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Martin Picard
- Department of Psychiatry, Division of Behavioral Medicine, Department of Neurology and CTNI, Columbia University Medical Centre, New York, NY, USA
| | - Heidi M McBride
- Neuromuscular Group, Department of Neurology and Neurosurgery, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC, Canada
| | - Yan Burelle
- Faculty of Pharmacy, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
29
|
Methylmercury alters glutathione homeostasis by inhibiting glutaredoxin 1 and enhancing glutathione biosynthesis in cultured human astrocytoma cells. Toxicol Lett 2016; 256:1-10. [PMID: 27180086 DOI: 10.1016/j.toxlet.2016.05.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 04/28/2016] [Accepted: 05/11/2016] [Indexed: 02/08/2023]
Abstract
Methylmercury (MeHg) is a neurotoxin that binds strongly to thiol residues on protein and low molecular weight molecules like reduced glutathione (GSH). The mechanism of its effects on GSH homeostasis particularly at environmentally relevant low doses is not fully known. We hypothesized that exposure to MeHg would lead to a depletion of reduced glutathione (GSH) and an accumulation of glutathione disulfide (GSSG) leading to alterations in S-glutathionylation of proteins. Our results showed exposure to low concentrations of MeHg (1μM) did not significantly alter GSH levels but increased GSSG levels by ∼12-fold. This effect was associated with a significant increase in total cellular glutathione content and a decrease in GSH/GSSG. Immunoblot analyses revealed that proteins involved in glutathione synthesis were upregulated accounting for the increase in cellular glutathione. This was associated an increase in cellular Nrf2 protein levels which is required to induce the expression of antioxidant genes in response to cellular stress. Intriguingly, we noted that a key enzyme involved in reversing protein S-glutathionylation and maintaining glutathione homeostasis, glutaredoxin-1 (Grx1), was inhibited by ∼50%. MeHg treatment also increased the S-glutathionylation of a high molecular weight protein. This observation is consistent with the inhibition of Grx1 and elevated H2O2 production however; contrary to our original hypothesis we found few S-glutathionylated proteins in the astrocytoma cells. Collectively, MeHg affects multiple arms of glutathione homeostasis ranging from pool management to protein S-glutathionylation and Grx1 activity.
Collapse
|
30
|
Mitochondrial Redox Signaling and Tumor Progression. Cancers (Basel) 2016; 8:cancers8040040. [PMID: 27023612 PMCID: PMC4846849 DOI: 10.3390/cancers8040040] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2015] [Revised: 02/21/2016] [Accepted: 03/07/2016] [Indexed: 01/10/2023] Open
Abstract
Cancer cell can reprogram their energy production by switching mitochondrial oxidative phosphorylation to glycolysis. However, mitochondria play multiple roles in cancer cells, including redox regulation, reactive oxygen species (ROS) generation, and apoptotic signaling. Moreover, these mitochondrial roles are integrated via multiple interconnected metabolic and redox sensitive pathways. Interestingly, mitochondrial redox proteins biphasically regulate tumor progression depending on cellular ROS levels. Low level of ROS functions as signaling messengers promoting cancer cell proliferation and cancer invasion. However, anti-cancer drug-initiated stress signaling could induce excessive ROS, which is detrimental to cancer cells. Mitochondrial redox proteins could scavenger basal ROS and function as “tumor suppressors” or prevent excessive ROS to act as “tumor promoter”. Paradoxically, excessive ROS often also induce DNA mutations and/or promotes tumor metastasis at various stages of cancer progression. Targeting redox-sensitive pathways and transcriptional factors in the appropriate context offers great promise for cancer prevention and therapy. However, the therapeutics should be cancer-type and stage-dependent.
Collapse
|
31
|
Mailloux RJ, Craig Ayre D, Christian SL. Induction of mitochondrial reactive oxygen species production by GSH mediated S-glutathionylation of 2-oxoglutarate dehydrogenase. Redox Biol 2016; 8:285-97. [PMID: 26928132 PMCID: PMC4776629 DOI: 10.1016/j.redox.2016.02.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 02/01/2016] [Accepted: 02/07/2016] [Indexed: 12/18/2022] Open
Abstract
2-Oxoglutarate dehydrogenase (Ogdh) is an important mitochondria redox sensor that can undergo S-glutathionylation following an increase in H2O2 levels. Although S-glutathionylation is required to protect Ogdh from irreversible oxidation while simultaneously modulating its activity it remains unknown if glutathione can also modulate reactive oxygen species (ROS) production by the complex. We report that reduced (GSH) and oxidized (GSSG) glutathione control O2∙-/H2O2 formation by Ogdh through protein S-glutathionylation reactions. GSSG (1 mM) induced a modest decrease in Ogdh activity which was associated with a significant decrease in O2∙-/H2O2 formation. GSH had the opposite effect, amplifying O2∙-/H2O2 formation by Ogdh. Incubation of purified Ogdh in 2.5 mM GSH led to significant increase in O2∙-/H2O2 formation which also lowered NADH production. Inclusion of enzymatically active glutaredoxin-2 (Grx2) in reaction mixtures reversed the GSH-mediated amplification of O2∙-/H2O2 formation. Similarly pre-incubation of permeabilized liver mitochondria from mouse depleted of GSH showed an approximately ~3.5-fold increase in Ogdh-mediated O2∙-/H2O2 production that was matched by a significant decrease in NADH formation which could be reversed by Grx2. Taken together, our results demonstrate GSH and GSSG modulate ROS production by Ogdh through S-glutathionylation of different subunits. This is also the first demonstration that GSH can work in the opposite direction in mitochondria-amplifying ROS formation instead of quenching it. We propose that this regulatory mechanism is required to modulate ROS emission from Ogdh in response to variations in glutathione redox buffering capacity. ROS formation by Ogdh is controlled by glutathione. GSH amplifies ROS production by Ogdh. Ogdh is S-glutathionylated by GSH. Grx2 deglutathionylates Ogdh.
Collapse
Affiliation(s)
- Ryan J Mailloux
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada.
| | - D Craig Ayre
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| | - Sherri L Christian
- Department of Biochemistry, Memorial University of Newfoundland, St. John's, Newfoundland, Canada
| |
Collapse
|
32
|
Yoshioka J. Thioredoxin superfamily and its effects on cardiac physiology and pathology. Compr Physiol 2016; 5:513-30. [PMID: 25880503 DOI: 10.1002/cphy.c140042] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A precise control of oxidation/reduction of protein thiols is essential for intact cardiac physiology. Irreversible oxidative modifications have been proposed to play a role in the pathogenesis of cardiovascular diseases. An imbalance of redox homeostasis with diminution of antioxidant capacities predisposes the heart to oxidant injury. There is growing interest in endoplasmic reticulum (ER) stress in the cardiovascular field, since perturbation of redox homeostasis in the ER is sufficient to cause ER stress. Because a number of human diseases are related to altered redox homeostasis and defects in protein folding, many research efforts have been devoted in recent years to understanding the structure and enzymatic properties of the thioredoxin superfamily. The thioredoxin superfamily has been well documented as thiol oxidoreductases to exert a role in various cell signaling pathways. The redox properties of the thioredoxin motif account for the different functions of several members of the thioredoxin superfamily. While thioredoxin and glutaredoxin primarily act as antioxidants by reducing protein disulfides and mixed disulfide, another member of the superfamily, protein disulfide isomerase (PDI), can act as an oxidant by forming intrachain disulfide bonds that contribute to proper protein folding. Increasing evidence suggests a pivotal role of PDI in the survival pathway that promotes cardiomyocyte survival and leads to more favorable cardiac remodeling. Thus, the thiol redox state is important for cellular redox signaling and survival pathway in the heart. This review summarizes the key features of major members of the thioredoxin superfamily directly involved in cardiac physiology and pathology.
Collapse
Affiliation(s)
- Jun Yoshioka
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Cambridge, Massachusetts, USA
| |
Collapse
|
33
|
Kramer PA, Duan J, Qian WJ, Marcinek DJ. The Measurement of Reversible Redox Dependent Post-translational Modifications and Their Regulation of Mitochondrial and Skeletal Muscle Function. Front Physiol 2015; 6:347. [PMID: 26635632 PMCID: PMC4658434 DOI: 10.3389/fphys.2015.00347] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Accepted: 11/09/2015] [Indexed: 12/28/2022] Open
Abstract
Mitochondrial oxidative stress is a common feature of skeletal myopathies across multiple conditions; however, the mechanism by which it contributes to skeletal muscle dysfunction remains controversial. Oxidative damage to proteins, lipids, and DNA has received the most attention, yet an important role for reversible redox post-translational modifications (PTMs) in pathophysiology is emerging. The possibility that these PTMs can exert dynamic control of muscle function implicates them as a mechanism contributing to skeletal muscle dysfunction in chronic disease. Herein, we discuss the significance of thiol-based redox dependent modifications to mitochondrial, myofibrillar, and excitation-contraction (EC) coupling proteins with an emphasis on how these changes could alter skeletal muscle performance under chronically stressed conditions. A major barrier to a better mechanistic understanding of the role of reversible redox PTMs in muscle function is the technical challenges associated with accurately measuring the changes of site-specific redox PTMs. Here we will critically review current approaches with an emphasis on sample preparation artifacts, quantitation, and specificity. Despite these challenges, the ability to accurately quantify reversible redox PTMs is critical to understanding the mechanisms by which mitochondrial oxidative stress contributes to skeletal muscle dysfunction in chronic diseases.
Collapse
Affiliation(s)
- Philip A Kramer
- Department of Radiology, University of Washington Seattle, WA, USA
| | - Jicheng Duan
- Biological Sciences Division, Pacific Northwest National Laboratory Richland, WA, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Pacific Northwest National Laboratory Richland, WA, USA
| | - David J Marcinek
- Department of Radiology, University of Washington Seattle, WA, USA ; Department of Bioengineering, University of Washington Seattle, WA, USA
| |
Collapse
|
34
|
Zamora DA, Downs KP, Ullevig SL, Tavakoli S, Kim HS, Qiao M, Greaves DR, Asmis R. Glutaredoxin 2a overexpression in macrophages promotes mitochondrial dysfunction but has little or no effect on atherogenesis in LDL-receptor null mice. Atherosclerosis 2015; 241:69-78. [PMID: 25966442 PMCID: PMC4466159 DOI: 10.1016/j.atherosclerosis.2015.04.805] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2014] [Revised: 04/23/2015] [Accepted: 04/24/2015] [Indexed: 12/31/2022]
Abstract
AIMS Reactive oxygen species (ROS)-mediated formation of mixed disulfides between critical cysteine residues in proteins and glutathione, a process referred to as protein S-glutathionylation, can lead to loss of enzymatic activity and protein degradation. Since mitochondria are a major source of ROS and a number of their proteins are susceptible to protein-S-glutathionylation, we examined if overexpression of mitochondrial thioltranferase glutaredoxin 2a (Grx2a) in macrophages of dyslipidemic atherosclerosis-prone mice would prevent mitochondrial dysfunction and protect against atherosclerotic lesion formation. METHODS AND RESULTS We generated transgenic Grx2aMac(LDLR-/-) mice, which overexpress Grx2a as an EGFP fusion protein under the control of the macrophage-specific CD68 promoter. Transgenic mice and wild type siblings were fed a high fat diet for 14 weeks at which time we assessed mitochondrial bioenergetic function in peritoneal macrophages and atherosclerotic lesion formation. Flow cytometry and Western blot analysis demonstrated transgene expression in blood monocytes and peritoneal macrophages isolated from Grx2aMac(LDLR-/-) mice, and fluorescence confocal microscopy studies confirmed that Grx2a expression was restricted to the mitochondria of monocytic cells. Live-cell bioenergetic measurements revealed impaired mitochondrial ATP turnover in macrophages isolated from Grx2aMac(LDLR-/-) mice compared to macrophages isolated from non-transgenic mice. However, despite impaired mitochondrial function in macrophages of Grx2aMac(LDLR-/-) mice, we observed no significant difference in the severity of atherosclerosis between wildtype and Grx2aMac(LDLR-/-) mice. CONCLUSION Our findings suggest that increasing Grx2a activity in macrophage mitochondria disrupts mitochondrial respiration and ATP production, but without affecting the proatherogenic potential of macrophages. Our data suggest that macrophages are resistant against moderate mitochondrial dysfunction and rely on alternative pathways for ATP synthesis to support the energetic requirements.
Collapse
Affiliation(s)
- D A Zamora
- Department of Biology, Trinity University, San Antonio, USA
| | - K P Downs
- Department of Clinical Laboratory Sciences, University of Texas Health Science Center at San Antonio, USA
| | - S L Ullevig
- Department of Kinesiology, Health, and Nutrition, University of Texas at San Antonio, San Antonio, USA
| | - S Tavakoli
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, USA
| | - H S Kim
- Department of Clinical Laboratory Sciences, University of Texas Health Science Center at San Antonio, USA
| | - M Qiao
- Department of Clinical Laboratory Sciences, University of Texas Health Science Center at San Antonio, USA
| | - D R Greaves
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - R Asmis
- Department of Clinical Laboratory Sciences, University of Texas Health Science Center at San Antonio, USA; Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, USA; Department of Biochemistry, University of Texas Health Science Center at San Antonio, USA.
| |
Collapse
|
35
|
Victorino VJ, Mencalha AL, Panis C. Post-translational modifications disclose a dual role for redox stress in cardiovascular pathophysiology. Life Sci 2015; 129:42-7. [DOI: 10.1016/j.lfs.2014.11.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2014] [Revised: 11/03/2014] [Accepted: 11/11/2014] [Indexed: 02/07/2023]
|
36
|
Mailloux RJ. Teaching the fundamentals of electron transfer reactions in mitochondria and the production and detection of reactive oxygen species. Redox Biol 2015; 4:381-98. [PMID: 25744690 PMCID: PMC4348434 DOI: 10.1016/j.redox.2015.02.001] [Citation(s) in RCA: 191] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/01/2015] [Indexed: 12/11/2022] Open
Abstract
Mitochondria fulfill a number of biological functions which inherently depend on ATP and O2(-•)/H2O2 production. Both ATP and O2(-•)/H2O2 are generated by electron transfer reactions. ATP is the product of oxidative phosphorylation whereas O2(-•) is generated by singlet electron reduction of di-oxygen (O2). O2(-•) is then rapidly dismutated by superoxide dismutase (SOD) producing H2O2. O2(-•)/H2O2 were once viewed as unfortunately by-products of aerobic respiration. This characterization is fitting considering over production of O2(-•)/H2O2 by mitochondria is associated with range of pathological conditions and aging. However, O2(-•)/H2O2 are only dangerous in large quantities. If produced in a controlled fashion and maintained at a low concentration, cells can benefit greatly from the redox properties of O2(-•)/H2O2. Indeed, low rates of O2(-•)/H2O2 production are required for intrinsic mitochondrial signaling (e.g. modulation of mitochondrial processes) and communication with the rest of the cell. O2(-•)/H2O2 levels are kept in check by anti-oxidant defense systems that sequester O2(-•)/H2O2 with extreme efficiency. Given the importance of O2(-•)/H2O2 in cellular function, it is imperative to consider how mitochondria produce O2(-•)/H2O2 and how O2(-•)/H2O2 genesis is regulated in conjunction with fluctuations in nutritional and redox states. Here, I discuss the fundamentals of electron transfer reactions in mitochondria and emerging knowledge on the 11 potential sources of mitochondrial O2(-•)/H2O2 in tandem with their significance in contributing to overall O2(-•)/H2O2 emission in health and disease. The potential for classifying these different sites in isopotential groups, which is essentially defined by the redox properties of electron donator involved in O2(-•)/H2O2 production, as originally suggested by Brand and colleagues is also surveyed in detail. In addition, redox signaling mechanisms that control O2(-•)/H2O2 genesis from these sites are discussed. Finally, the current methodologies utilized for measuring O2(-•)/H2O2 in isolated mitochondria, cell culture and in vivo are reviewed.
Collapse
Affiliation(s)
- Ryan J Mailloux
- Department of Biology, Faculty of Sciences, University of Ottawa, 30 Marie Curie, Ottawa, ON, Canada K1N 6N5.
| |
Collapse
|
37
|
Mailloux RJ, Willmore WG. S-glutathionylation reactions in mitochondrial function and disease. Front Cell Dev Biol 2014; 2:68. [PMID: 25453035 PMCID: PMC4233936 DOI: 10.3389/fcell.2014.00068] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2014] [Accepted: 10/31/2014] [Indexed: 01/23/2023] Open
Abstract
Mitochondria are highly efficient energy-transforming organelles that convert energy stored in nutrients into ATP. The production of ATP by mitochondria is dependent on oxidation of nutrients and coupling of exergonic electron transfer reactions to the genesis of transmembrane electrochemical potential of protons. Electrons can also prematurely “spin-off” from prosthetic groups in Krebs cycle enzymes and respiratory complexes and univalently reduce di-oxygen to generate reactive oxygen species (ROS) superoxide (O2•−) and hydrogen peroxide (H2O2), important signaling molecules that can be toxic at high concentrations. Production of ATP and ROS are intimately linked by the respiratory chain and the genesis of one or the other inherently depends on the metabolic state of mitochondria. Various control mechanisms converge on mitochondria to adjust ATP and ROS output in response to changing cellular demands. One control mechanism that has gained a high amount of attention recently is S-glutathionylation, a redox sensitive covalent modification that involves formation of a disulfide bridge between glutathione and an available protein cysteine thiol. A number of S-glutathionylation targets have been identified in mitochondria. It has also been established that S-glutathionylation reactions in mitochondria are mediated by the thiol oxidoreductase glutaredoxin-2 (Grx2). In the following review, emerging knowledge on S-glutathionylation reactions and its importance in modulating mitochondrial ATP and ROS production will be discussed. Major focus will be placed on Complex I of the respiratory chain since (1) it is a target for reversible S-glutathionylation by Grx2 and (2) deregulation of Complex I S-glutathionylation is associated with development of various disease states particularly heart disease. Other mitochondrial enzymes and how their S-glutathionylation profile is affected in different disease states will also be discussed.
Collapse
Affiliation(s)
- Ryan J Mailloux
- Department of Biology, Faculty of Sciences, University of Ottawa Ottawa, ON, Canada
| | | |
Collapse
|
38
|
Canonical and new generation anticancer drugs also target energy metabolism. Arch Toxicol 2014; 88:1327-50. [PMID: 24792321 DOI: 10.1007/s00204-014-1246-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Accepted: 04/15/2014] [Indexed: 01/05/2023]
Abstract
Significant efforts have been made for the development of new anticancer drugs (protein kinase or proteasome inhibitors, monoclonal humanized antibodies) with presumably low or negligible side effects and high specificity. However, an in-depth analysis of the side effects of several currently used canonical (platin-based drugs, taxanes, anthracyclines, etoposides, antimetabolites) and new generation anticancer drugs as the first line of clinical treatment reveals significant perturbation of glycolysis and oxidative phosphorylation. Canonical and new generation drug side effects include decreased (1) intracellular ATP levels, (2) glycolytic/mitochondrial enzyme/transporter activities and/or (3) mitochondrial electrical membrane potentials. Furthermore, the anti-proliferative effects of these drugs are markedly attenuated in tumor rho (0) cells, in which functional mitochondria are absent; in addition, several anticancer drugs directly interact with isolated mitochondria affecting their functions. Therefore, several anticancer drugs also target the energy metabolism, and hence, the documented inhibitory effect of anticancer drugs on cancer growth should also be linked to the blocking of ATP supply pathways. These often overlooked effects of canonical and new generation anticancer drugs emphasize the role of energy metabolism in maintaining cancer cells viable and its targeting as a complementary and successful strategy for cancer treatment.
Collapse
|
39
|
Mailloux RJ, Xuan JY, McBride S, Maharsy W, Thorn S, Holterman CE, Kennedy CRJ, Rippstein P, deKemp R, da Silva J, Nemer M, Lou M, Harper ME. Glutaredoxin-2 is required to control oxidative phosphorylation in cardiac muscle by mediating deglutathionylation reactions. J Biol Chem 2014; 289:14812-28. [PMID: 24727547 DOI: 10.1074/jbc.m114.550574] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Glutaredoxin-2 (Grx2) modulates the activity of several mitochondrial proteins in cardiac tissue by catalyzing deglutathionylation reactions. However, it remains uncertain whether Grx2 is required to control mitochondrial ATP output in heart. Here, we report that Grx2 plays a vital role modulating mitochondrial energetics and heart physiology by mediating the deglutathionylation of mitochondrial proteins. Deletion of Grx2 (Grx2(-/-)) decreased ATP production by complex I-linked substrates to half that in wild type (WT) mitochondria. Decreased respiration was associated with increased complex I glutathionylation diminishing its activity. Tissue glucose uptake was concomitantly increased. Mitochondrial ATP output and complex I activity could be recovered by restoring the redox environment to that favoring the deglutathionylated states of proteins. Grx2(-/-) hearts also developed left ventricular hypertrophy and fibrosis, and mice became hypertensive. Mitochondrial energetics from Grx2 heterozygotes (Grx2(+/-)) were also dysfunctional, and hearts were hypertrophic. Intriguingly, Grx2(+/-) mice were far less hypertensive than Grx2(-/-) mice. Thus, Grx2 plays a vital role in modulating mitochondrial metabolism in cardiac muscle, and Grx2 deficiency leads to pathology. As mitochondrial ATP production was restored by the addition of reductants, these findings may be relevant to novel redox-related therapies in cardiac disease.
Collapse
Affiliation(s)
- Ryan J Mailloux
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Jian Ying Xuan
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Skye McBride
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Wael Maharsy
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Stephanie Thorn
- the University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Chet E Holterman
- the Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada, and
| | - Christopher R J Kennedy
- the Kidney Research Centre, Ottawa Hospital Research Institute, Ottawa Hospital, Ottawa, Ontario K1H 8L6, Canada, and
| | - Peter Rippstein
- the University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Robert deKemp
- the University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Jean da Silva
- the University of Ottawa Heart Institute, Ottawa, Ontario K1Y 4W7, Canada
| | - Mona Nemer
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada
| | - Marjorie Lou
- the Center of Redox Biology and School of Veterinary Medicine and Biomedical Sciences, University of Nebraska at Lincoln, Lincoln, Nebraska 68583-0903
| | - Mary-Ellen Harper
- From the Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario K1H 8M5, Canada,
| |
Collapse
|
40
|
Bei Y, Hong P. A novel approach to minimize false discovery rate in genome-wide data analysis. BMC SYSTEMS BIOLOGY 2014; 7 Suppl 4:S1. [PMID: 24564975 PMCID: PMC3856609 DOI: 10.1186/1752-0509-7-s4-s1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
BACKGROUND High-throughput technologies, such as DNA microarray, have significantly advanced biological and biomedical research by enabling researchers to carry out genome-wide screens. One critical task in analyzing genome-wide datasets is to control the false discovery rate (FDR) so that the proportion of false positive features among those called significant is restrained. Recently a number of FDR control methods have been proposed and widely practiced, such as the Benjamini-Hochberg approach, the Storey approach and Significant Analysis of Microarrays (SAM). METHODS This paper presents a straight-forward yet powerful FDR control method termed miFDR, which aims to minimize FDR when calling a fixed number of significant features. We theoretically proved that the strategy used by miFDR is able to find the optimal number of significant features when the desired FDR is fixed. RESULTS We compared miFDR with the BH approach, the Storey approach and SAM on both simulated datasets and public DNA microarray datasets. The results demonstrated that miFDR outperforms others by identifying more significant features under the same FDR cut-offs. Literature search showed that many genes called only by miFDR are indeed relevant to the underlying biology of interest. CONCLUSIONS FDR has been widely applied to analyzing high-throughput datasets allowed for rapid discoveries. Under the same FDR threshold, miFDR is capable to identify more significant features than its competitors at a compatible level of complexity. Therefore, it can potentially generate great impacts on biological and biomedical research. AVAILABILITY If interested, please contact the authors for getting miFDR.
Collapse
|
41
|
Lian X, Wang H, Wei X, Wang Y, Wang Q, Guo L, Zhao Y, Chen X. BMI‑1 is important in bufalin‑induced apoptosis of K562 cells. Mol Med Rep 2014; 9:1209-17. [PMID: 24566825 DOI: 10.3892/mmr.2014.1980] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Accepted: 01/23/2014] [Indexed: 11/06/2022] Open
Abstract
The purpose of this study was to analyze the effects of bufalin on the gene expression of K562 cells and on the expression of BMI‑1 pathway constituents in K562 cell apoptosis. K562 cells were treated with bufalin, and the inhibition rate and apoptosis were detected by an MTT assay, flow cytometry and a microarray assay. BMI‑1, p16INK4a and p14ARF were examined by quantitative polymerase chain reaction (qPCR). Bufalin induced significant changes in the gene expression of the K562 cells; 4296 genes were differentially expressed, 2185 were upregulated and 2111 were downregulated. The most upregulated genes were associated with transcription regulation, while the most downregulated genes were associated with the non-coding RNA metabolic processes and DNA repair. qPCR analysis demonstrated that BMI‑1 was overexpressed in the K562 cells. Bufalin is able to downregulate BMI‑1 expression levels in K562 cells prematurely and cause an increase in the expression levels of p16INK4a and p14ARF. Moreover, bufalin downregulated BCR/ABL expression levels in a time‑dependent manner, and the expression of BCR/ABL was not associated with the upregulation or downregulation of BMI‑1 expression. Bufalin may induce K562 cell apoptosis by downregulating BMI‑1 expression levels and accordingly upregulating the expression levels of p16INK4a and p14ARF. Bufalin may also induce K562 cell apoptosis via downregulating BCR/ABL expression levels, and this pathway may be independent of the BMI‑1 pathway.
Collapse
Affiliation(s)
- Xiaoyun Lian
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| | - Hao Wang
- Department of Hematology, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Xucang Wei
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Yi Wang
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Qishan Wang
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Liang Guo
- Department of Hematology, Xi'an Central Hospital, Xi'an, Shaanxi 710003, P.R. China
| | - Yuan Zhao
- Department of Hematology, Shaanxi Provincial People's Hospital, Xi'an, Shaanxi 710068, P.R. China
| | - Xiequn Chen
- Department of Hematology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, P.R. China
| |
Collapse
|
42
|
Lagoa R, Gañán C, López-Sánchez C, García-Martínez V, Gutierrez-Merino C. The decrease of NAD(P)H:quinone oxidoreductase 1 activity and increase of ROS production by NADPH oxidases are early biomarkers in doxorubicin cardiotoxicity. Biomarkers 2014; 19:142-53. [PMID: 24506563 DOI: 10.3109/1354750x.2014.885084] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CONTEXT Doxorubicin cardiotoxicity displays a complex and multifactorial progression. OBJECTIVE Identify early biochemical mechanisms leading to a sustained imbalance of cellular bioenergetics. METHODS Measurements of the temporal evolution of selected biochemical markers after treatment of rats with doxorubicin (20 mg/kg body weight). RESULTS Doxorubicin treatment increased lipid oxidation, catalase activity and production of H₂O₂ by Nox-NADPH oxidases, and down-regulated NAD(P)H quinone oxidoreductase-1 prior eliciting changes in reduced glutathione, protein carbonyls and protein nitrotyrosines. Alterations of mitochondrial and myofibrillar bioenergetics biomarkers were detected only after this oxidative imbalance was established. CONCLUSIONS NAD(P)H quinone oxidoreductase-1 activity and increase of hydrogen peroxide production by NADPH oxidases are early biomarkers in doxorubicin cardiotoxicity.
Collapse
Affiliation(s)
- Ricardo Lagoa
- ESTG-Polytechnic Institute of Leiria , Leiria , Portugal
| | | | | | | | | |
Collapse
|
43
|
Mailloux RJ, Jin X, Willmore WG. Redox regulation of mitochondrial function with emphasis on cysteine oxidation reactions. Redox Biol 2013; 2:123-39. [PMID: 24455476 PMCID: PMC3895620 DOI: 10.1016/j.redox.2013.12.011] [Citation(s) in RCA: 210] [Impact Index Per Article: 19.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Accepted: 12/13/2013] [Indexed: 12/13/2022] Open
Abstract
Mitochondria have a myriad of essential functions including metabolism and apoptosis. These chief functions are reliant on electron transfer reactions and the production of ATP and reactive oxygen species (ROS). The production of ATP and ROS are intimately linked to the electron transport chain (ETC). Electrons from nutrients are passed through the ETC via a series of acceptor and donor molecules to the terminal electron acceptor molecular oxygen (O2) which ultimately drives the synthesis of ATP. Electron transfer through the respiratory chain and nutrient oxidation also produces ROS. At high enough concentrations ROS can activate mitochondrial apoptotic machinery which ultimately leads to cell death. However, if maintained at low enough concentrations ROS can serve as important signaling molecules. Various regulatory mechanisms converge upon mitochondria to modulate ATP synthesis and ROS production. Given that mitochondrial function depends on redox reactions, it is important to consider how redox signals modulate mitochondrial processes. Here, we provide the first comprehensive review on how redox signals mediated through cysteine oxidation, namely S-oxidation (sulfenylation, sulfinylation), S-glutathionylation, and S-nitrosylation, regulate key mitochondrial functions including nutrient oxidation, oxidative phosphorylation, ROS production, mitochondrial permeability transition (MPT), apoptosis, and mitochondrial fission and fusion. We also consider the chemistry behind these reactions and how they are modulated in mitochondria. In addition, we also discuss emerging knowledge on disorders and disease states that are associated with deregulated redox signaling in mitochondria and how mitochondria-targeted medicines can be utilized to restore mitochondrial redox signaling.
Collapse
Affiliation(s)
- Ryan J. Mailloux
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, Canada K1S 5B6
- Toxicology Research Division, Food Directorate, HPFB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - Xiaolei Jin
- Toxicology Research Division, Food Directorate, HPFB, Health Canada, Ottawa, Ontario, Canada K1A 0K9
| | - William G. Willmore
- Institute of Biochemistry, Carleton University, 1125 Colonel By Drive, Ottawa, Ontario, Canada K1S 5B6
| |
Collapse
|
44
|
Gao XH, Qanungo S, Pai HV, Starke DW, Steller KM, Fujioka H, Lesnefsky EJ, Kerner J, Rosca MG, Hoppel CL, Mieyal JJ. Aging-dependent changes in rat heart mitochondrial glutaredoxins--Implications for redox regulation. Redox Biol 2013; 1:586-98. [PMID: 25126518 PMCID: PMC4127417 DOI: 10.1016/j.redox.2013.10.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 10/28/2013] [Accepted: 10/29/2013] [Indexed: 12/17/2022] Open
Abstract
Clinical and animal studies have documented that hearts of the elderly are more susceptible to ischemia/reperfusion damage compared to young adults. Recently we found that aging-dependent increase in susceptibility of cardiomyocytes to apoptosis was attributable to decrease in cytosolic glutaredoxin 1 (Grx1) and concomitant decrease in NF-κB-mediated expression of anti-apoptotic proteins. Besides primary localization in the cytosol, Grx1 also exists in the mitochondrial intermembrane space (IMS). In contrast, Grx2 is confined to the mitochondrial matrix. Here we report that Grx1 is decreased by 50–60% in the IMS, but Grx2 is increased by 1.4–2.6 fold in the matrix of heart mitochondria from elderly rats. Determination of in situ activities of the Grx isozymes from both subsarcolemmal (SSM) and interfibrillar (IFM) mitochondria revealed that Grx1 was fully active in the IMS. However, Grx2 was mostly in an inactive form in the matrix, consistent with reversible sequestration of the active-site cysteines of two Grx2 molecules in complex with an iron–sulfur cluster. Our quantitative evaluations of the active/inactive ratio for Grx2 suggest that levels of dimeric Grx2 complex with iron–sulfur clusters are increased in SSM and IFM in the hearts of elderly rats. We found that the inactive Grx2 can be fully reactivated by sodium dithionite or exogenous superoxide production mediated by xanthine oxidase. However, treatment with rotenone, which generates intramitochondrial superoxide through inhibition of mitochondrial respiratory chain Complex I, did not lead to Grx2 activation. These findings suggest that insufficient ROS accumulates in the vicinity of dimeric Grx2 to activate it in situ. Glutaredoxins play key roles in cellular redox regulation, which is sensitive to aging-dependent dysregulation. Grx1 is diminished in the intermembrane space of mitochondria from aged heart; matrix Grx2 is increased but mostly in an inactive form. The inactive Grx2 is selectively activated by superoxide. Mitochondrial glutaredoxin changes may contribute to dysregulation of redox homeostasis during aging. Changes in in situ activities of heart mitochondrial Grx1 and Grx2 with aging provide mechanistic insights for future studies.
Collapse
Key Words
- Aging
- Cys-SSG, l-cysteine–glutathione mixed disulfide
- DT, sodium dithionite
- GSH, reduced glutathione
- GSSG, glutathione disulfide
- Glutaredoxin
- Glutathionylation
- Grx, glutaredoxin
- IFM, Heart interfibrillar mitochondria
- Iron–sulfur cluster
- Mitochondria
- Mn-TMPyP, Mn(III) tetrakis (1-methyl-4-pyridyl) porphyrin
- Reactive oxygen species (ROS)
- Redox regulation
- SSM, heart subsarcolemmal mitochondria
- t-Bid, caspase-8-cleaved human BID
- tetratosylate, hydroxide
Collapse
Affiliation(s)
- Xing-Huang Gao
- Department of Pharmacology, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Suparna Qanungo
- Department of Pharmacology, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Harish V Pai
- Department of Pharmacology, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - David W Starke
- Department of Pharmacology, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Kelly M Steller
- Department of Pharmacology, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA ; Louis Stokes Cleveland Veterans Affairs Medical Research Center, Cleveland, OH 44106, USA
| | - Hisashi Fujioka
- Center for Mitochondrial Disease, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Edward J Lesnefsky
- Department of Medicine, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Janos Kerner
- Department of Pharmacology, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA ; Center for Mitochondrial Disease, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Mariana G Rosca
- Department of Pharmacology, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA ; Center for Mitochondrial Disease, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - Charles L Hoppel
- Department of Pharmacology, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA ; Center for Mitochondrial Disease, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA ; Department of Medicine, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA
| | - John J Mieyal
- Department of Pharmacology, Division of Cardiology, Case Western Reserve University, School of Medicine, Cleveland, OH 44106, USA ; Louis Stokes Cleveland Veterans Affairs Medical Research Center, Cleveland, OH 44106, USA
| |
Collapse
|
45
|
Hanschmann EM, Godoy JR, Berndt C, Hudemann C, Lillig CH. Thioredoxins, glutaredoxins, and peroxiredoxins--molecular mechanisms and health significance: from cofactors to antioxidants to redox signaling. Antioxid Redox Signal 2013; 19:1539-605. [PMID: 23397885 PMCID: PMC3797455 DOI: 10.1089/ars.2012.4599] [Citation(s) in RCA: 496] [Impact Index Per Article: 45.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 02/01/2013] [Accepted: 02/07/2013] [Indexed: 12/19/2022]
Abstract
Thioredoxins (Trxs), glutaredoxins (Grxs), and peroxiredoxins (Prxs) have been characterized as electron donors, guards of the intracellular redox state, and "antioxidants". Today, these redox catalysts are increasingly recognized for their specific role in redox signaling. The number of publications published on the functions of these proteins continues to increase exponentially. The field is experiencing an exciting transformation, from looking at a general redox homeostasis and the pathological oxidative stress model to realizing redox changes as a part of localized, rapid, specific, and reversible redox-regulated signaling events. This review summarizes the almost 50 years of research on these proteins, focusing primarily on data from vertebrates and mammals. The role of Trx fold proteins in redox signaling is discussed by looking at reaction mechanisms, reversible oxidative post-translational modifications of proteins, and characterized interaction partners. On the basis of this analysis, the specific regulatory functions are exemplified for the cellular processes of apoptosis, proliferation, and iron metabolism. The importance of Trxs, Grxs, and Prxs for human health is addressed in the second part of this review, that is, their potential impact and functions in different cell types, tissues, and various pathological conditions.
Collapse
Affiliation(s)
- Eva-Maria Hanschmann
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, Ernst-Moritz Arndt University, Greifswald, Germany
| | - José Rodrigo Godoy
- Institute of Physiology, Pathophysiology and Biophysics, Department of Biomedical Sciences, University of Veterinary Medicine, Vienna, Austria
| | - Carsten Berndt
- Department of Neurology, Medical Faculty, Heinrich-Heine University, Duesseldorf, Germany
| | - Christoph Hudemann
- Institute of Laboratory Medicine, Molecular Diagnostics, Philipps University, Marburg, Germany
| | - Christopher Horst Lillig
- Institute for Medical Biochemistry and Molecular Biology, University Medicine, Ernst-Moritz Arndt University, Greifswald, Germany
| |
Collapse
|
46
|
Mailloux RJ, McBride SL, Harper ME. Unearthing the secrets of mitochondrial ROS and glutathione in bioenergetics. Trends Biochem Sci 2013; 38:592-602. [PMID: 24120033 DOI: 10.1016/j.tibs.2013.09.001] [Citation(s) in RCA: 203] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 09/04/2013] [Accepted: 09/05/2013] [Indexed: 01/06/2023]
Abstract
During the cellular oxidation of fuels, electrons are used to power the proton pumps of the mitochondrial electron transport chain (ETC) and ultimately drive ATP synthesis and the reduction of molecular oxygen to water. During these oxidative processes, some electrons can 'spin off' during fuel oxidation and electron transport to univalently reduce O2, forming reactive oxygen species (ROS). In excess, ROS can be detrimental; however, at low concentrations oxyradicals are essential signaling molecules. Mitochondria thus use a battery of systems to finely control types and levels of ROS, including antioxidants. Several antioxidant systems depend on glutathione. Here, we review mitochondrial ROS homeostatic systems, including emerging knowledge about roles of glutathione in redox balance and the control of protein function by post-translational modification.
Collapse
Affiliation(s)
- Ryan J Mailloux
- Department of Biochemistry, Microbiology, and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | | | | |
Collapse
|
47
|
Gupta A, Rohlfsen C, Leppo MK, Chacko VP, Wang Y, Steenbergen C, Weiss RG. Creatine kinase-overexpression improves myocardial energetics, contractile dysfunction and survival in murine doxorubicin cardiotoxicity. PLoS One 2013; 8:e74675. [PMID: 24098344 PMCID: PMC3788056 DOI: 10.1371/journal.pone.0074675] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/05/2013] [Indexed: 11/19/2022] Open
Abstract
Doxorubicin (DOX) is a commonly used life-saving antineoplastic agent that also causes dose-dependent cardiotoxicity. Because ATP is absolutely required to sustain normal cardiac contractile function and because impaired ATP synthesis through creatine kinase (CK), the primary myocardial energy reserve reaction, may contribute to contractile dysfunction in heart failure, we hypothesized that impaired CK energy metabolism contributes to DOX-induced cardiotoxicity. We therefore overexpressed the myofibrillar isoform of CK (CK-M) in the heart and determined the energetic, contractile and survival effects of CK-M following weekly DOX (5mg/kg) administration using in vivo31P MRS and 1H MRI. In control animals, in vivo cardiac energetics were reduced at 7 weeks of DOX protocol and this was followed by a mild but significant reduction in left ventricular ejection fraction (EF) at 8 weeks of DOX, as compared to baseline. At baseline, CK-M overexpression (CK-M-OE) increased rates of ATP synthesis through cardiac CK (CK flux) but did not affect contractile function. Following DOX however, CK-M-OE hearts had better preservation of creatine phosphate and higher CK flux and higher EF as compared to control DOX hearts. Survival after DOX administration was significantly better in CK-M-OE than in control animals (p<0.02). Thus CK-M-OE attenuates the early decline in myocardial high-energy phosphates and contractile function caused by chronic DOX administration and increases survival. These findings suggest that CK impairment plays an energetic and functional role in this DOX-cardiotoxicity model and suggests that metabolic strategies, particularly those targeting CK, offer an appealing new strategy for limiting DOX-associated cardiotoxicity.
Collapse
Affiliation(s)
- Ashish Gupta
- Department of Medicine, Division of Cardiology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Radiology, Division of Magnetic Resonance Research, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Cory Rohlfsen
- Department of Medicine, Division of Cardiology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Michelle K. Leppo
- Department of Medicine, Division of Cardiology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Vadappuram P. Chacko
- Department of Radiology, Division of Magnetic Resonance Research, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Yibin Wang
- University of California Los Angeles, Los Angeles, California, United States of America
| | - Charles Steenbergen
- Department of Pathology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Robert G. Weiss
- Department of Medicine, Division of Cardiology, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Department of Radiology, Division of Magnetic Resonance Research, the Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
48
|
Pang X, Panee J, Liu X, Berry MJ, Chang SL, Chang L. Regional variations of antioxidant capacity and oxidative stress responses in HIV-1 transgenic rats with and without methamphetamine administration. J Neuroimmune Pharmacol 2013; 8:691-704. [PMID: 23546885 PMCID: PMC3773562 DOI: 10.1007/s11481-013-9454-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/17/2013] [Indexed: 11/24/2022]
Abstract
HIV infection and methamphetamine (Meth) abuse both may lead to oxidative stress. This study used HIV-1 transgenic (HIV-1Tg) rats to investigate the independent and combined effects of HIV viral protein expression and low dose repeated Meth exposure on the glutathione (GSH)-centered antioxidant system and oxidative stress in the brain. Total GSH content, gene expression and/or enzymatic activities of glutamylcysteine synthetase (GCS), gamma-glutamyltransferase (GGT), glutathione reductase (GR), glutathione peroxidase (GPx), glutaredoxin (Glrx), and glutathione-s-transferase (GST) were measured. The protein expression of cystine transporter (xCT) and oxidative stress marker 4-hydroxynonenal (HNE) were also analyzed. Brain regions studied include thalamus, frontal and remainder cortex, striatum, cerebellum and hippocampus. HIV-1Tg rats and Meth exposure showed highly regional specific responses. In the F344 rats, the thalamus had the highest baseline GSH concentration and potentially higher GSH recycle rate. HIV-1Tg rats showed strong transcriptional responses to GSH depletion in the thalamus. Both HIV-1Tg and Meth resulted in decreased GR activity in thalamus, and decreased Glrx activity in frontal cortex. However, the increased GR and Glrx activities synergized with increased GSH concentration, which might have partially prevented Meth-induced oxidative stress in striatum. Interactive effects between Meth and HIV-1Tg were observed in thalamus on the activities of GCS and GGT, and in thalamus and frontal cortex on Glrx activity and xCT protein expression. Findings suggest that HIV viral protein and low dose repeated Meth exposure have separate and combined effects on the brain's antioxidant capacity and the oxidative stress response that are regional specific.
Collapse
Affiliation(s)
- Xiaosha Pang
- Department of Cell and Molecular Biology, John A. Burns
School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street BSB 222,
Honolulu HI 96813
| | - Jun Panee
- Department of Cell and Molecular Biology, John A. Burns
School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street BSB 222,
Honolulu HI 96813
| | - Xiangqian Liu
- Institute of NeuroImmune Pharmacology and Department of
Biological Sciences, Seton Hall University, South Orange, NJ 07079
- Department of Histology and Embryology, Tongji Medical
College, Huazhong University of Science and Technology, Wuhan 430030, P.R.
China
| | - Marla J. Berry
- Department of Cell and Molecular Biology, John A. Burns
School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street BSB 222,
Honolulu HI 96813
| | - Sulie L. Chang
- Institute of NeuroImmune Pharmacology and Department of
Biological Sciences, Seton Hall University, South Orange, NJ 07079
| | - Linda Chang
- Department of Cell and Molecular Biology, John A. Burns
School of Medicine, University of Hawaii at Manoa, 651 Ilalo Street BSB 222,
Honolulu HI 96813
| |
Collapse
|
49
|
Ghezzi P. Protein glutathionylation in health and disease. Biochim Biophys Acta Gen Subj 2013; 1830:3165-72. [DOI: 10.1016/j.bbagen.2013.02.009] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2012] [Revised: 01/10/2013] [Accepted: 02/07/2013] [Indexed: 12/31/2022]
|
50
|
Handy DE, Loscalzo J. Redox regulation of mitochondrial function. Antioxid Redox Signal 2012; 16:1323-67. [PMID: 22146081 PMCID: PMC3324814 DOI: 10.1089/ars.2011.4123] [Citation(s) in RCA: 372] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 12/06/2011] [Accepted: 12/06/2011] [Indexed: 02/06/2023]
Abstract
Redox-dependent processes influence most cellular functions, such as differentiation, proliferation, and apoptosis. Mitochondria are at the center of these processes, as mitochondria both generate reactive oxygen species (ROS) that drive redox-sensitive events and respond to ROS-mediated changes in the cellular redox state. In this review, we examine the regulation of cellular ROS, their modes of production and removal, and the redox-sensitive targets that are modified by their flux. In particular, we focus on the actions of redox-sensitive targets that alter mitochondrial function and the role of these redox modifications on metabolism, mitochondrial biogenesis, receptor-mediated signaling, and apoptotic pathways. We also consider the role of mitochondria in modulating these pathways, and discuss how redox-dependent events may contribute to pathobiology by altering mitochondrial function.
Collapse
Affiliation(s)
- Diane E Handy
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|