1
|
Zhang C, Chang X, Zhao D, He Y, Dong G, Gao L. Mitochondria and myocardial ischemia/reperfusion injury: Effects of Chinese herbal medicine and the underlying mechanisms. J Pharm Anal 2025; 15:101051. [PMID: 39931135 PMCID: PMC11808734 DOI: 10.1016/j.jpha.2024.101051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 07/12/2024] [Accepted: 07/19/2024] [Indexed: 02/03/2025] Open
Abstract
Ischemic heart disease (IHD) is associated with high morbidity and mortality rates. Reperfusion therapy is the best treatment option for this condition. However, reperfusion can aggravate myocardial damage through a phenomenon known as myocardial ischemia/reperfusion (I/R) injury, which has recently gained the attention of researchers. Several studies have shown that Chinese herbal medicines and their natural monomeric components exert therapeutic effects against I/R injury. This review outlines the current knowledge on the pathological mechanisms through which mitochondria participate in I/R injury, focusing on the issues related to energy metabolism, mitochondrial quality control disorders, oxidative stress, and calcium. The mechanisms by which mitochondria mediate cell death have also been discussed. To develop a resource for the prevention and management of clinical myocardial I/R damage, we compiled the most recent research on the effects of Chinese herbal remedies and their monomer components.
Collapse
Affiliation(s)
- Chuxin Zhang
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xing Chang
- Guang'anmen Hospital of Chinese Academy of Traditional Chinese Medicine, Beijing, 100053, China
| | - Dandan Zhao
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yu He
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Guangtong Dong
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Lin Gao
- Beijing University of Chinese Medicine, Beijing, 100029, China
| |
Collapse
|
2
|
Martinez CS, Zheng A, Xiao Q. Mitochondrial Reactive Oxygen Species Dysregulation in Heart Failure with Preserved Ejection Fraction: A Fraction of the Whole. Antioxidants (Basel) 2024; 13:1330. [PMID: 39594472 PMCID: PMC11591317 DOI: 10.3390/antiox13111330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a multifarious syndrome, accounting for over half of heart failure (HF) patients receiving clinical treatment. The prevalence of HFpEF is rapidly increasing in the coming decades as the global population ages. It is becoming clearer that HFpEF has a lot of different causes, which makes it challenging to find effective treatments. Currently, there are no proven treatments for people with deteriorating HF or HFpEF. Although the pathophysiologic foundations of HFpEF are complex, excessive reactive oxygen species (ROS) generation and increased oxidative stress caused by mitochondrial dysfunction seem to play a critical role in the pathogenesis of HFpEF. Emerging evidence from animal models and human myocardial tissues from failed hearts shows that mitochondrial aberrations cause a marked increase in mitochondrial ROS (mtROS) production and oxidative stress. Furthermore, studies have reported that common HF medications like beta blockers, angiotensin receptor blockers, angiotensin-converting enzyme inhibitors, and mineralocorticoid receptor antagonists indirectly reduce the production of mtROS. Despite the harmful effects of ROS on cardiac remodeling, maintaining mitochondrial homeostasis and cardiac functions requires small amounts of ROS. In this review, we will provide an overview and discussion of the recent findings on mtROS production, its threshold for imbalance, and the subsequent dysfunction that leads to related cardiac and systemic phenotypes in the context of HFpEF. We will also focus on newly discovered cellular and molecular mechanisms underlying ROS dysregulation, current therapeutic options, and future perspectives for treating HFpEF by targeting mtROS and the associated signal molecules.
Collapse
Affiliation(s)
| | | | - Qingzhong Xiao
- Centre for Clinical Pharmacology and Precision Medicine, William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK; (C.S.M.); (A.Z.)
| |
Collapse
|
3
|
Zhang S, Liu Z, Zhang H, Zhou X, Wang X, Chen Y, Miao X, Zhu Y, Jiang W. Effect and mechanism of Qing Gan Zi Shen decoction on heart damage induced by obesity and hypertension. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117163. [PMID: 37741474 DOI: 10.1016/j.jep.2023.117163] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 08/15/2023] [Accepted: 09/07/2023] [Indexed: 09/25/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Qing Gan Zi Shen Decoction (QGZS) is a traditional Chinese formula. It has been extensively used for decades in the treatment of hypertension combined with metabolic diseases, but its cardioprotective effects and underlying mechanisms are poorly understood. AIM OF THE STUDY To explore the cardioprotective effects and potential mechanisms of QGZS in an animal model of obese hypertension. MATERIALS AND METHODS In this study, spontaneously hypertensive rats (SHRs) were utilized as an animal model to examine the effects of a high-fat diet and two concentrations of QGZS. Echocardiography, hematoxylin eosin (H&E) staining, and wheat germ agglutinin (WGA) staining were employed to assess the cardiac structure and function of the SHRs throughout a 16-week therapy period. Furthermore, Western blotting (WB) and immunofluorescence (IF) were employed to identify the levels of Nrf2 expression in the mitochondria, cytoplasm, and nucleus of the myocardium. Additionally, transmission electron microscopy and enzyme-linked immunosorbent assay (ELISA) were utilized to measure mitochondrial morphology and pro-inflammatory cytokine levels, respectively. Furthermore, Western blotting (WB), immunohistochemistry (IHC), and immunofluorescence (IF) techniques were employed to quantify the levels of marker proteins associated with myocardial fibrosis, cardiac inflammation, oxidative stress, and mitochondrial dysfunction. RESULTS QGZS inhibited weight gain and depressed systolic and mean arterial pressures in high-fat-fed SHRs. Echocardiographic results demonstrated that QGZS prevented the increase in left ventricular mass, restricted the growth of left ventricular diameter, and improved ejection fraction (EF), fractional shortening (FS), and the ratio of early diastolic peak velocity of transmitral flow (E) to late diastolic peak velocity (A) in high-fat-fed SHRs. This suggested that QGZS prevented ventricular remodeling and protected cardiac systolic and diastolic functions. H&E and WGA staining showed that QGZS improved cardiomyocyte disorders and restricted cardiomyocyte hypertrophy. The underlying mechanisms, QGZS attenuated the oxidative stress state, including reducing the generation of reactive oxygen species (ROS) in the myocardium, revitalizing the antioxidant enzyme system, and protecting mitochondrial function. Moreover, QGZS alleviated the pro-inflammatory state in high-fat-fed SHRs. What's more, QGZS significantly increased the expression level of Nrf2 in nuclei and mitochondria in rat heart tissues, exerting a proximate Nrf2 agonist effect. CONCLUSIONS QGZS exerted cardioprotective effects, in part due to its increasing expression of Nrf2 protein in the heart, which promoted Nrf2 nuclear expression.
Collapse
Affiliation(s)
- Shujie Zhang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Zitian Liu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Han Zhang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Xiaonian Zhou
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Xiuming Wang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Yan Chen
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Xiaofan Miao
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China
| | - Yao Zhu
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China.
| | - Weimin Jiang
- Department of Cardiology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, 210023, Jiangsu, PR China.
| |
Collapse
|
4
|
Heusch G, Andreadou I, Bell R, Bertero E, Botker HE, Davidson SM, Downey J, Eaton P, Ferdinandy P, Gersh BJ, Giacca M, Hausenloy DJ, Ibanez B, Krieg T, Maack C, Schulz R, Sellke F, Shah AM, Thiele H, Yellon DM, Di Lisa F. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol 2023; 67:102894. [PMID: 37839355 PMCID: PMC10590874 DOI: 10.1016/j.redox.2023.102894] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The present review summarizes the beneficial and detrimental roles of reactive oxygen species in myocardial ischemia/reperfusion injury and cardioprotection. In the first part, the continued need for cardioprotection beyond that by rapid reperfusion of acute myocardial infarction is emphasized. Then, pathomechanisms of myocardial ischemia/reperfusion to the myocardium and the coronary circulation and the different modes of cell death in myocardial infarction are characterized. Different mechanical and pharmacological interventions to protect the ischemic/reperfused myocardium in elective percutaneous coronary interventions and coronary artery bypass grafting, in acute myocardial infarction and in cardiotoxicity from cancer therapy are detailed. The second part keeps the focus on ROS providing a comprehensive overview of molecular and cellular mechanisms involved in ischemia/reperfusion injury. Starting from mitochondria as the main sources and targets of ROS in ischemic/reperfused myocardium, a complex network of cellular and extracellular processes is discussed, including relationships with Ca2+ homeostasis, thiol group redox balance, hydrogen sulfide modulation, cross-talk with NAPDH oxidases, exosomes, cytokines and growth factors. While mechanistic insights are needed to improve our current therapeutic approaches, advancements in knowledge of ROS-mediated processes indicate that detrimental facets of oxidative stress are opposed by ROS requirement for physiological and protective reactions. This inevitable contrast is likely to underlie unsuccessful clinical trials and limits the development of novel cardioprotective interventions simply based upon ROS removal.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert Bell
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Edoardo Bertero
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genova, Genova, Italy
| | - Hans-Erik Botker
- Department of Cardiology, Institute for Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - James Downey
- Department of Physiology, University of South Alabama, Mobile, AL, USA
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Heart Centre, Charterhouse Square, London, United Kingdom
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, National Heart Research Institute Singapore, National Heart Centre, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), IIS-Fundación Jiménez Díaz University Hospital, and CIBERCV, Madrid, Spain
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig -Universität, Giessen, Germany
| | - Frank Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Fabio Di Lisa
- Dipartimento di Scienze Biomediche, Università degli studi di Padova, Padova, Italy.
| |
Collapse
|
5
|
Kaludercic N, Arusei RJ, Di Lisa F. Recent advances on the role of monoamine oxidases in cardiac pathophysiology. Basic Res Cardiol 2023; 118:41. [PMID: 37792081 PMCID: PMC10550854 DOI: 10.1007/s00395-023-01012-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/17/2023] [Accepted: 09/18/2023] [Indexed: 10/05/2023]
Abstract
Numerous physiological and pathological roles have been attributed to the formation of mitochondrial reactive oxygen species (ROS). However, the individual contribution of different mitochondrial processes independently of bioenergetics remains elusive and clinical treatments unavailable. A notable exception to this complexity is found in the case of monoamine oxidases (MAOs). Unlike other ROS-producing enzymes, especially within mitochondria, MAOs possess a distinct combination of defined molecular structure, substrate specificity, and clinically accessible inhibitors. Another significant aspect of MAO activity is the simultaneous generation of hydrogen peroxide alongside highly reactive aldehydes and ammonia. These three products synergistically impair mitochondrial function at various levels, ultimately jeopardizing cellular metabolic integrity and viability. This pathological condition arises from exacerbated MAO activity, observed in many cardiovascular diseases, thus justifying the exploration of MAO inhibitors as effective cardioprotective strategy. In this context, we not only summarize the deleterious roles of MAOs in cardiac pathologies and the positive effects resulting from genetic or pharmacological MAO inhibition, but also discuss recent findings that expand our understanding on the role of MAO in gene expression and cardiac development.
Collapse
Affiliation(s)
- Nina Kaludercic
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy.
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35127, Padua, Italy.
| | - Ruth Jepchirchir Arusei
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Via Ugo Bassi 58/B, 35131, Padua, Italy.
- Neuroscience Institute, National Research Council of Italy (CNR), 35131, Padua, Italy.
| |
Collapse
|
6
|
Hernansanz-Agustín P, Enríquez JA. Alternative respiratory oxidases to study the animal electron transport chain. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2023; 1864:148936. [PMID: 36395975 DOI: 10.1016/j.bbabio.2022.148936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 10/05/2022] [Accepted: 11/06/2022] [Indexed: 11/16/2022]
Abstract
Oxidative phosphorylation is a common process to most organisms in which the main function is to generate an electrochemical gradient across the inner mitochondrial membrane (IMM) and to make energy available to the cell. However, plants, many fungi and some animals maintain non-energy conserving oxidases which serve as a bypass to coupled respiration. Namely, the alternative NADH:ubiquinone oxidoreductase NDI1, present in the complex I (CI)-lacking Saccharomyces cerevisiae, and the alternative oxidase, ubiquinol:oxygen oxidoreductase AOX, present in many organisms across different kingdoms. In the last few years, these alternative oxidases have been used to dissect previously indivisible processes in bioenergetics and have helped to discover, understand, and corroborate important processes in mitochondria. Here, we review how the use of alternative oxidases have contributed to the knowledge in CI stability, bioenergetics, redox biology, and the implications of their use in current and future research.
Collapse
Affiliation(s)
- Pablo Hernansanz-Agustín
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; Centro de Investigaciones Biomédicas en Red en Fragilidad y Envejecimiento saludable (CIBERFES), 28029 Madrid, Spain.
| | - José Antonio Enríquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain; Centro de Investigaciones Biomédicas en Red en Fragilidad y Envejecimiento saludable (CIBERFES), 28029 Madrid, Spain.
| |
Collapse
|
7
|
P66Shc (Shc1) Zebrafish Mutant Line as a Platform for Testing Decreased Reactive Oxygen Species in Pathology. J Cardiovasc Dev Dis 2022; 9:jcdd9110385. [DOI: 10.3390/jcdd9110385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 11/01/2022] [Accepted: 11/02/2022] [Indexed: 11/12/2022] Open
Abstract
Reactive oxygen species (ROS) dysregulation exacerbates many pathologies but must remain within normal ranges to maintain cell function. Since ROS-mediated pathology and routine cell function are coupled, in vivo models evaluating low-ROS background effects on pathology are limited. Some models alter enzymatic antioxidant expression/activity, while others involve small molecule antioxidant administration. These models cause non-specific ROS neutralization, decreasing both beneficial and detrimental ROS. This is detrimental in cardiovascular pathology, despite the negative effects excessive ROS has on these pathologies. Thus, current trends in ROS-mediated pathology have shifted toward selective inhibition of ROS producers that are dysregulated during pathological insults, such as p66Shc. In this study, we evaluated a zebrafish heterozygote p66Shc hypomorphic mutant line as a low-ROS myocardial infarction (MI) pathology model that mimics mammalian MI. Our findings suggest this zebrafish line does not have an associated negative phenotype, but has decreased body mass and tissue ROS levels that confer protection against ROS-mediated pathology. Therefore, this line may provide a low-ROS background leading to new insights into disease.
Collapse
|
8
|
Prag HA, Aksentijevic D, Dannhorn A, Giles AV, Mulvey JF, Sauchanka O, Du L, Bates G, Reinhold J, Kula-Alwar D, Xu Z, Pellerin L, Goodwin RJA, Murphy MP, Krieg T. Ischemia-Selective Cardioprotection by Malonate for Ischemia/Reperfusion Injury. Circ Res 2022; 131:528-541. [PMID: 35959683 PMCID: PMC9426742 DOI: 10.1161/circresaha.121.320717] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
BACKGROUND Inhibiting SDH (succinate dehydrogenase), with the competitive inhibitor malonate, has shown promise in ameliorating ischemia/reperfusion injury. However, key for translation to the clinic is understanding the mechanism of malonate entry into cells to enable inhibition of SDH, its mitochondrial target, as malonate itself poorly permeates cellular membranes. The possibility of malonate selectively entering the at-risk heart tissue on reperfusion, however, remains unexplored. METHODS C57BL/6J mice, C2C12 and H9c2 myoblasts, and HeLa cells were used to elucidate the mechanism of selective malonate uptake into the ischemic heart upon reperfusion. Cells were treated with malonate while varying pH or together with transport inhibitors. Mouse hearts were either perfused ex vivo (Langendorff) or subjected to in vivo left anterior descending coronary artery ligation as models of ischemia/reperfusion injury. Succinate and malonate levels were assessed by liquid chromatography-tandem mass spectrometry LC-MS/MS, in vivo by mass spectrometry imaging, and infarct size by TTC (2,3,5-triphenyl-2H-tetrazolium chloride) staining. RESULTS Malonate was robustly protective against cardiac ischemia/reperfusion injury, but only if administered at reperfusion and not when infused before ischemia. The extent of malonate uptake into the heart was proportional to the duration of ischemia. Malonate entry into cardiomyocytes in vivo and in vitro was dramatically increased at the low pH (≈6.5) associated with ischemia. This increased uptake of malonate was blocked by selective inhibition of MCT1 (monocarboxylate transporter 1). Reperfusion of the ischemic heart region with malonate led to selective SDH inhibition in the at-risk region. Acid-formulation greatly enhances the cardioprotective potency of malonate. CONCLUSIONS Cardioprotection by malonate is dependent on its entry into cardiomyocytes. This is facilitated by the local decrease in pH that occurs during ischemia, leading to its selective uptake upon reperfusion into the at-risk tissue, via MCT1. Thus, malonate's preferential uptake in reperfused tissue means it is an at-risk tissue-selective drug that protects against cardiac ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Hiran A. Prag
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom.,MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom
| | - Dunja Aksentijevic
- Centre for Biochemical Pharmacology, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, United Kingdom (D.A.)
| | - Andreas Dannhorn
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom (A.D., R.J.A.G.)
| | - Abigail V. Giles
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom.,MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom.,Laboratory of Cardiac Energetics, National Heart, Lung and Blood Institute, Bethesda, MD (A.V.G.)
| | - John F. Mulvey
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| | - Olga Sauchanka
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| | - Luping Du
- Department of Physiology and Pathophysiology, Tianjin Medical University, China (L.D., Z.X.)
| | - Georgina Bates
- MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom
| | - Johannes Reinhold
- MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom.,Faculty of Medicine and Health Sciences, University of East Anglia, Norwich Research Park (J.R.)
| | - Duvaraka Kula-Alwar
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| | - Zhelong Xu
- Department of Physiology and Pathophysiology, Tianjin Medical University, China (L.D., Z.X.)
| | - Luc Pellerin
- Département de Physiologie, Université de Lausanne, Switzerland (L.P.).,Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS, LabEx TRAIL-IBIO, Université de Bordeaux, France (L.P.).,Inserm U1313, Université et CHU de Poitiers, France (L.P.)
| | - Richard J. A. Goodwin
- Imaging and Data Analytics, Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, United Kingdom (A.D., R.J.A.G.).,Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom (R.J.A.G.)
| | - Michael P. Murphy
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom.,MRC Mitochondrial Biology Unit (H.A.P., A.V.G., G.B., J.R., M.M.P.), University of Cambridge, United Kingdom
| | - Thomas Krieg
- Department of Medicine (H.A.P., A.V.G., J.F.M., O.S., D.K.-A., M.P.M., T.K.), University of Cambridge, United Kingdom
| |
Collapse
|
9
|
Pande S, Raisuddin S. The Underexplored Dimensions of Nutritional Hormesis. Curr Nutr Rep 2022; 11:386-394. [PMID: 35723856 DOI: 10.1007/s13668-022-00423-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/23/2022] [Indexed: 10/18/2022]
Abstract
PURPOSE OF REVIEW Hormesis is biphasic response wherein low and high doses of chemical and nutrient confer beneficial and toxic effects respectively, typically in a U-shaped manner. Hormesis is intricately related to bioenergetic state of a cell, and therefore, nutrition impacts it. Excessive nutrition can halt the endogenous antioxidant synthesis leading to cytotoxic effects. While low and optimum doses of the same bring about hormetic stimulation that can exalt the antioxidant response and reduce susceptibility towards degenerative diseases. The sirtuin family of proteins is triggered by mild stress of calorie restriction and exerts hormesis. Similarly, several phytochemicals and micronutrients are known to bring about health benefits at optimum dose and deleterious effects at high doses. Despite this attribute, nutritional hormesis is not very well researched upon because the magnitude of hormetic effect observed is generally quite modest. There is no precise regulation of optimal intake of certain foods to witness hormesis and no characterization of any biomarker that reports stress responses at various doses above or below optimal intakes. There is a major gap in research between nutrition and hormesis being affected by sirtuin family of proteins, phytochemicals, and micronutrients. RECENT FINDINGS Mild stress of calorie restriction elevates sirtuin protein and effect of sirtuin protein on hormesis has been recently reported. More foods that enhance sirtuin protein, phytochemicals, and micronutrients need to be explored in relation to hormesis and associated health benefits.
Collapse
Affiliation(s)
- Shubhra Pande
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India.
| | - Sheikh Raisuddin
- Department of Medical Elementology and Toxicology, Jamia Hamdard (Hamdard University), New Delhi, India
| |
Collapse
|
10
|
Bellini C, Antonucci S, Morillas-Becerril L, Scarpa S, Tavano R, Mancin F, Di Lisa F, Papini E. Nanoparticles Based on Cross-Linked Poly(Lipoic Acid) Protect Macrophages and Cardiomyocytes from Oxidative Stress and Ischemia Reperfusion Injury. Antioxidants (Basel) 2022; 11:antiox11050907. [PMID: 35624771 PMCID: PMC9137738 DOI: 10.3390/antiox11050907] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 04/29/2022] [Accepted: 05/02/2022] [Indexed: 12/10/2022] Open
Abstract
The control of radical damage and oxidative stress, phenomena involved in a large number of human pathologies, is a major pharmaceutical and medical goal. We here show that two biocompatible formulations of Pluronic-stabilized, poly (lipoic acid)-based nanoparticles (NP) effectively antagonized the formation of radicals and reactive oxygen species (ROS). These NPs, not only intrinsically scavenged radicals in a-cellular DPPH/ABTS assays, but also inhibited the overproduction of ROS induced by tert-Butyl hydroperoxide (t-BHP) in tumor cells (HeLa), human macrophages and neonatal rat ventricular myocytes (NRVMs). NPs were captured by macrophages and cardiomyocytes much more effectively as compared to HeLa cells and non-phagocytic leukocytes, eventually undergoing intracellular disassembly. Notably, NPs decreased the mitochondrial ROS generation induced by simulated Ischemia/Reperfusion Injury (IRI) in isolated cardiomyocytes. NPs also prevented IRI-triggered cardiomyocyte necrosis, mitochondrial dysfunction, and alterations of contraction-related intracellular Ca2+ waves. Hence, NPs appear to be an effective and cardiomyocyte-selective drug to protect against damages induced by post-ischemic reperfusion.
Collapse
Affiliation(s)
- Chiara Bellini
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy; (C.B.); (S.A.); (S.S.); (R.T.)
- CRIBI—Centre for Innovative Biotechnology Research, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy
| | - Salvatore Antonucci
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy; (C.B.); (S.A.); (S.S.); (R.T.)
| | - Lucía Morillas-Becerril
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35121 Padova, Italy; (L.M.-B.); (F.M.)
| | - Sara Scarpa
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy; (C.B.); (S.A.); (S.S.); (R.T.)
- CRIBI—Centre for Innovative Biotechnology Research, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy
| | - Regina Tavano
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy; (C.B.); (S.A.); (S.S.); (R.T.)
- CRIBI—Centre for Innovative Biotechnology Research, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy
| | - Fabrizio Mancin
- Department of Chemical Sciences, University of Padova, Via F. Marzolo 1, 35121 Padova, Italy; (L.M.-B.); (F.M.)
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy; (C.B.); (S.A.); (S.S.); (R.T.)
- Correspondence: (F.D.L.); (E.P.)
| | - Emanuele Papini
- Department of Biomedical Sciences, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy; (C.B.); (S.A.); (S.S.); (R.T.)
- CRIBI—Centre for Innovative Biotechnology Research, University of Padova, Via U. Bassi 58/b, 35121 Padova, Italy
- Correspondence: (F.D.L.); (E.P.)
| |
Collapse
|
11
|
Mallik B, Frank CA. Roles for Mitochondrial Complex I Subunits in Regulating Synaptic Transmission and Growth. Front Neurosci 2022; 16:846425. [PMID: 35557603 PMCID: PMC9087048 DOI: 10.3389/fnins.2022.846425] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/22/2022] [Indexed: 11/13/2022] Open
Abstract
To identify conserved components of synapse function that are also associated with human diseases, we conducted a genetic screen. We used the Drosophila melanogaster neuromuscular junction (NMJ) as a model. We employed RNA interference (RNAi) on selected targets and assayed synapse function and plasticity by electrophysiology. We focused our screen on genetic factors known to be conserved from human neurological or muscle functions (300 Drosophila lines screened). From our screen, knockdown of a Mitochondrial Complex I (MCI) subunit gene (ND-20L) lowered levels of NMJ neurotransmission. Due to the severity of the phenotype, we studied MCI function further. Knockdown of core MCI subunits concurrently in neurons and muscle led to impaired neurotransmission. We localized this neurotransmission function to the muscle. Pharmacology targeting MCI phenocopied the impaired neurotransmission phenotype. Finally, MCI subunit knockdowns or pharmacological inhibition led to profound cytological defects, including reduced NMJ growth and altered NMJ morphology. Mitochondria are essential for cellular bioenergetics and produce ATP through oxidative phosphorylation. Five multi-protein complexes achieve this task, and MCI is the largest. Impaired Mitochondrial Complex I subunits in humans are associated with disorders such as Parkinson’s disease, Leigh syndrome, and cardiomyopathy. Together, our data present an analysis of Complex I in the context of synapse function and plasticity. We speculate that in the context of human MCI dysfunction, similar neuronal and synaptic defects could contribute to pathogenesis.
Collapse
Affiliation(s)
- Bhagaban Mallik
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, United States
| | - C. Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA, United States
- Carver College of Medicine and Iowa Neuroscience Institute, University of Iowa, Iowa City, IA, United States
- *Correspondence: C. Andrew Frank,
| |
Collapse
|
12
|
Burger N, James AM, Mulvey JF, Hoogewijs K, Ding S, Fearnley IM, Loureiro-López M, Norman AAI, Arndt S, Mottahedin A, Sauchanka O, Hartley RC, Krieg T, Murphy MP. ND3 Cys39 in complex I is exposed during mitochondrial respiration. Cell Chem Biol 2022; 29:636-649.e14. [PMID: 34739852 PMCID: PMC9076552 DOI: 10.1016/j.chembiol.2021.10.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2021] [Revised: 07/21/2021] [Accepted: 10/07/2021] [Indexed: 12/13/2022]
Abstract
Mammalian complex I can adopt catalytically active (A-) or deactive (D-) states. A defining feature of the reversible transition between these two defined states is thought to be exposure of the ND3 subunit Cys39 residue in the D-state and its occlusion in the A-state. As the catalytic A/D transition is important in health and disease, we set out to quantify it by measuring Cys39 exposure using isotopic labeling and mass spectrometry, in parallel with complex I NADH/CoQ oxidoreductase activity. To our surprise, we found significant Cys39 exposure during NADH/CoQ oxidoreductase activity. Furthermore, this activity was unaffected if Cys39 alkylation occurred during complex I-linked respiration. In contrast, alkylation of catalytically inactive complex I irreversibly blocked the reactivation of NADH/CoQ oxidoreductase activity by NADH. Thus, Cys39 of ND3 is exposed in complex I during mitochondrial respiration, with significant implications for our understanding of the A/D transition and the mechanism of complex I.
Collapse
Affiliation(s)
- Nils Burger
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Andrew M James
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - John F Mulvey
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Kurt Hoogewijs
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; The Wellcome Trust Centre for Mitochondrial Research, Institute for Cell and Molecular Biosciences, Newcastle University, Newcastle Upon Tyne NE2 4HH, UK; Medical Research Council-Laboratory of Molecular Biology, Cambridge CB2 0QH, UK
| | - Shujing Ding
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Ian M Fearnley
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Marta Loureiro-López
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | | | - Sabine Arndt
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Amin Mottahedin
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK; Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, 405 30 Gothenburg, Sweden
| | - Olga Sauchanka
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | | | - Thomas Krieg
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge CB2 0QQ, UK
| | - Michael P Murphy
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK.
| |
Collapse
|
13
|
Prag HA, Pala L, Kula-Alwar D, Mulvey JF, Luping D, Beach TE, Booty LM, Hall AR, Logan A, Sauchanka V, Caldwell ST, Robb EL, James AM, Xu Z, Saeb-Parsy K, Hartley RC, Murphy MP, Krieg T. Ester Prodrugs of Malonate with Enhanced Intracellular Delivery Protect Against Cardiac Ischemia-Reperfusion Injury In Vivo. Cardiovasc Drugs Ther 2022; 36:1-13. [PMID: 32648168 PMCID: PMC8770414 DOI: 10.1007/s10557-020-07033-6] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/26/2020] [Indexed: 12/01/2022]
Abstract
PURPOSE Mitochondrial reactive oxygen species (ROS) production upon reperfusion of ischemic tissue initiates the ischemia/reperfusion (I/R) injury associated with heart attack. During ischemia, succinate accumulates and its oxidation upon reperfusion by succinate dehydrogenase (SDH) drives ROS production. Inhibition of succinate accumulation and/or oxidation by dimethyl malonate (DMM), a cell permeable prodrug of the SDH inhibitor malonate, can decrease I/R injury. However, DMM is hydrolysed slowly, requiring administration to the heart prior to ischemia, precluding its administration to patients at the point of reperfusion, for example at the same time as unblocking a coronary artery following a heart attack. To accelerate malonate delivery, here we developed more rapidly hydrolysable malonate esters. METHODS We synthesised a series of malonate esters and assessed their uptake and hydrolysis by isolated mitochondria, C2C12 cells and in mice in vivo. In addition, we assessed protection against cardiac I/R injury by the esters using an in vivo mouse model of acute myocardial infarction. RESULTS We found that the diacetoxymethyl malonate diester (MAM) most rapidly delivered large amounts of malonate to cells in vivo. Furthermore, MAM could inhibit mitochondrial ROS production from succinate oxidation and was protective against I/R injury in vivo when added at reperfusion. CONCLUSIONS The rapidly hydrolysed malonate prodrug MAM can protect against cardiac I/R injury in a clinically relevant mouse model.
Collapse
Affiliation(s)
- Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Laura Pala
- School of Chemistry, University of Glasgow, Glasgow, G12 8QQ, UK
| | | | - John F Mulvey
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | - Du Luping
- Tianjin Medical University, Tianjin, 300070, China
| | - Timothy E Beach
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
| | - Lee M Booty
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Andrew R Hall
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Angela Logan
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Volha Sauchanka
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK
| | | | - Ellen L Robb
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Andrew M James
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Zhelong Xu
- Tianjin Medical University, Tianjin, 300070, China
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, CB2 0QQ, UK
| | | | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, CB2 0QQ, UK.
| |
Collapse
|
14
|
Prasad A, Mahmood A, Gupta R, Bisoyi P, Saleem N, Naga Prasad SV, Goswami SK. In cardiac muscle cells, both adrenergic agonists and antagonists induce reactive oxygen species from NOX2 but mutually attenuate each other's effects. Eur J Pharmacol 2021; 908:174350. [PMID: 34265295 DOI: 10.1016/j.ejphar.2021.174350] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/07/2021] [Accepted: 07/12/2021] [Indexed: 11/25/2022]
Abstract
In cardiac muscle cells adrenergic agonists stimulate the generation of reactive oxygen species, followed by redox signaling. We postulated that the antagonists would attenuate such reactive oxygen species generation by the agonists. H9c2 cardiac myoblasts, neonatal rat cardiac myocytes, and HEK293 cells expressing β1/β2 adrenoceptors were stimulated with several agonists and antagonists. All the agonists and antagonists independently generated reactive oxygen species; but its generation was minimum whenever an agonists was added together with an antagonist. We monitored the Ca++ signaling in the treated cells and obtained similar results. In all treatment sets, superoxide and H2O2 were generated in the mitochondria and the cytosol respectively. NOX2 inhibitor gp91ds-tat blocked reactive oxygen species generation by both the agonists and the antagonists. The level of p47phox subunit of NOX2 rapidly increased upon treatment, and it translocated to the plasma membrane, confirming NOX2 activation. Inhibitor studies showed that the activation of NOX2 involves ERK, PI3K, and tyrosine kinases. Recombinant promoter-reporter assays showed that reactive oxygen species generated by both the agonists and antagonists modulated downstream gene expression. Mice injected with the β-adrenergic agonist isoproterenol and fed with the antagonist metoprolol showed a robust induction of p47phox in the heart. We conclude that both the agonism and antagonism of adrenoceptors initiate redox signaling but when added together, they mutually counteract each other's effects. Our study thus highlights the importance of reactive oxygen species in adrenoceptor agonism and antagonism with relevance to the therapeutic use of the β blockers.
Collapse
Affiliation(s)
- Anamika Prasad
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
| | - Amena Mahmood
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India; DDU-Kaushal Kendra, Centre for Physiotherapy and Rehabilitation Sciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Richa Gupta
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
| | - Padmini Bisoyi
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
| | - Nikhat Saleem
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India
| | - Sathyamangla V Naga Prasad
- NB50, Department of Molecular Cardiology, Lerner Research Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH, 44195, USA.
| | - Shyamal K Goswami
- School of Life Sciences, Jawaharlal Nehru University, New Mehrauli Road, New Delhi, 110067, India.
| |
Collapse
|
15
|
Crewe C, Funcke JB, Li S, Joffin N, Gliniak CM, Ghaben AL, An YA, Sadek HA, Gordillo R, Akgul Y, Chen S, Samovski D, Fischer-Posovszky P, Kusminski CM, Klein S, Scherer PE. Extracellular vesicle-based interorgan transport of mitochondria from energetically stressed adipocytes. Cell Metab 2021; 33:1853-1868.e11. [PMID: 34418352 PMCID: PMC8429176 DOI: 10.1016/j.cmet.2021.08.002] [Citation(s) in RCA: 229] [Impact Index Per Article: 57.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Revised: 05/25/2021] [Accepted: 08/04/2021] [Indexed: 12/14/2022]
Abstract
Adipocytes undergo intense energetic stress in obesity resulting in loss of mitochondrial mass and function. We have found that adipocytes respond to mitochondrial stress by rapidly and robustly releasing small extracellular vesicles (sEVs). These sEVs contain respiration-competent, but oxidatively damaged mitochondrial particles, which enter circulation and are taken up by cardiomyocytes, where they trigger a burst of ROS. The result is compensatory antioxidant signaling in the heart that protects cardiomyocytes from acute oxidative stress, consistent with a preconditioning paradigm. As such, a single injection of sEVs from energetically stressed adipocytes limits cardiac ischemia/reperfusion injury in mice. This study provides the first description of functional mitochondrial transfer between tissues and the first vertebrate example of "inter-organ mitohormesis." Thus, these seemingly toxic adipocyte sEVs may provide a physiological avenue of potent cardio-protection against the inevitable lipotoxic or ischemic stresses elicited by obesity.
Collapse
Affiliation(s)
- Clair Crewe
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jan-Bernd Funcke
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shujuan Li
- Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Pediatric Cardiology, First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Nolwenn Joffin
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Christy M Gliniak
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexandra L Ghaben
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yu A An
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hesham A Sadek
- Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ruth Gordillo
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yucel Akgul
- Department of Plastic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Shiuhwei Chen
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Dmitri Samovski
- Center for Human Nutrition and the Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Pamela Fischer-Posovszky
- Department of Pediatrics and Adolescent Medicine, Ulm University Medical Center, 89075 Ulm, Germany
| | - Christine M Kusminski
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Samuel Klein
- Center for Human Nutrition and the Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Philipp E Scherer
- Touchstone Diabetes Center, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
16
|
Daiber A, Steven S, Euler G, Schulz R. Vascular and Cardiac Oxidative Stress and Inflammation as Targets for Cardioprotection. Curr Pharm Des 2021; 27:2112-2130. [PMID: 33550963 DOI: 10.2174/1381612827666210125155821] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 11/11/2020] [Indexed: 11/22/2022]
Abstract
Cardiac and vascular diseases are often associated with increased oxidative stress and inflammation, and both may contribute to the disease progression. However, successful applications of antioxidants in the clinical setting are very rare and specific anti-inflammatory therapeutics only emerged recently. Reasons for this rely on the great diversity of oxidative stress and inflammatory cells that can either act as cardioprotective or cause tissue damage in the heart. Recent large-scale clinical trials found that highly specific anti-inflammatory therapies using monoclonal antibodies against cytokines resulted in lower cardiovascular mortality in patients with pre-existing atherosclerotic disease. In addition, unspecific antiinflammatory medication and established cardiovascular drugs with pleiotropic immunomodulatory properties such as angiotensin converting enzyme (ACE) inhibitors or statins have proven beneficial cardiovascular effects. Normalization of oxidative stress seems to be a common feature of these therapies, which can be explained by a close interaction/crosstalk of the cellular redox state and inflammatory processes. In this review, we give an overview of cardiac reactive oxygen species (ROS) sources and processes of cardiac inflammation as well as the connection of ROS and inflammation in ischemic cardiomyopathy in order to shed light on possible cardioprotective interventions.
Collapse
Affiliation(s)
- Andreas Daiber
- Department of Cardiology, Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Sebastian Steven
- Department of Cardiology, Molecular Cardiology, University Medical Center Mainz, Mainz, Germany
| | - Gerhild Euler
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| | - Rainer Schulz
- Institute of Physiology, Justus-Liebig University, Giessen, Germany
| |
Collapse
|
17
|
Bou-Teen D, Kaludercic N, Weissman D, Turan B, Maack C, Di Lisa F, Ruiz-Meana M. Mitochondrial ROS and mitochondria-targeted antioxidants in the aged heart. Free Radic Biol Med 2021; 167:109-124. [PMID: 33716106 DOI: 10.1016/j.freeradbiomed.2021.02.043] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 02/14/2021] [Accepted: 02/26/2021] [Indexed: 12/12/2022]
Abstract
Excessive mitochondrial ROS production has been causally linked to the pathophysiology of aging in the heart and other organs, and plays a deleterious role in several age-related cardiac pathologies, including myocardial ischemia-reperfusion injury and heart failure, the two worldwide leading causes of death and disability in the elderly. However, ROS generation is also a fundamental mitochondrial function that orchestrates several signaling pathways, some of them exerting cardioprotective effects. In cardiac myocytes, mitochondria are particularly abundant and are specialized in subcellular populations, in part determined by their relationships with other organelles and their cyclic calcium handling activity necessary for adequate myocardial contraction/relaxation and redox balance. Depending on their subcellular location, mitochondria can themselves be differentially targeted by ROS and display distinct age-dependent functional decline. Thus, precise mitochondria-targeted therapies aimed at counteracting unregulated ROS production are expected to have therapeutic benefits in certain aging-related heart conditions. However, for an adequate design of such therapies, it is necessary to unravel the complex and dynamic interactions between mitochondria and other cellular processes.
Collapse
Affiliation(s)
- Diana Bou-Teen
- Hospital Universitari Vall d'Hebron, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR),Universitat Autonoma de Barcelona, 08035, Barcelona, Spain
| | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), via Ugo Bassi 58/B, 35131, Padova, Italy; Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), 35129, Padova, Italy
| | - David Weissman
- Comprehensive Heart Failure Center, University Clinic Würzburg, 97080, Würzburg, Germany
| | - Belma Turan
- Departments of Biophysics, Faculty of Medicine, Lokman Hekim University, Ankara, Turkey
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, 97080, Würzburg, Germany
| | - Fabio Di Lisa
- Neuroscience Institute, National Research Council of Italy (CNR), via Ugo Bassi 58/B, 35131, Padova, Italy; Department of Biomedical Sciences, University of Padova, via Ugo Bassi 58/B, 35131, Padova, Italy
| | - Marisol Ruiz-Meana
- Hospital Universitari Vall d'Hebron, Department of Cardiology, Vall d'Hebron Institut de Recerca (VHIR),Universitat Autonoma de Barcelona, 08035, Barcelona, Spain; Centro de Investigación Biomédica en Red-CV, CIBER-CV, Spain.
| |
Collapse
|
18
|
Xiao Y, Yim K, Zhang H, Bakker D, Nederlof R, Smeitink JAM, Renkema H, Hollmann MW, Weber NC, Zuurbier CJ. The Redox Modulating Sonlicromanol Active Metabolite KH176m and the Antioxidant MPG Protect Against Short-Duration Cardiac Ischemia-Reperfusion Injury. Cardiovasc Drugs Ther 2021; 35:745-758. [PMID: 33914182 PMCID: PMC8266721 DOI: 10.1007/s10557-021-07189-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/12/2021] [Indexed: 01/06/2023]
Abstract
Purpose Sonlicromanol is a phase IIB clinical stage compound developed for treatment of mitochondrial diseases. Its active component, KH176m, functions as an antioxidant, directly scavenging reactive oxygen species (ROS), and redox activator, boosting the peroxiredoxin-thioredoxin system. Here, we examined KH176m’s potential to protect against acute cardiac ischemia-reperfusion injury (IRI), compare it with the classic antioxidant N-(2-mercaptopropionyl)-glycine (MPG), and determine whether protection depends on duration (severity) of ischemia. Methods Isolated C56Bl/6N mouse hearts were Langendorff-perfused and subjected to short (20 min) or long (30 min) ischemia, followed by reperfusion. During perfusion, hearts were treated with saline, 10 μM KH176m, or 1 mM MPG. Cardiac function, cell death (necrosis), and mitochondrial damage (cytochrome c (CytC) release) were evaluated. In additional series, the effect of KH176m treatment on the irreversible oxidative stress marker 4-hydroxy-2-nonenal (4-HNE), formed during ischemia only, was determined at 30-min reperfusion. Results During baseline conditions, both drugs reduced cardiac performance, with opposing effects on vascular resistance (increased with KH176m, decreased with MPG). For short ischemia, KH176m robustly reduced all cell death parameters: LDH release (0.2 ± 0.2 vs 0.8 ± 0.5 U/min/GWW), infarct size (15 ± 8 vs 31 ± 20%), and CytC release (168.0 ± 151.9 vs 790.8 ± 453.6 ng/min/GWW). Protection by KH176m was associated with decreased cardiac 4-HNE. MPG only reduced CytC release. Following long ischemia, IRI was doubled, and KH176m and MPG now only reduced LDH release. The reduced protection against long ischemia was associated with the inability to reduce cardiac 4-HNE. Conclusion Protection against cardiac IRI by the antioxidant KH176m is critically dependent on duration of ischemia. The data suggest that with longer ischemia, the capacity of KH176m to reduce cardiac oxidative stress is rate-limiting, irreversible ischemic oxidative damage maximally accumulates, and antioxidant protection is strongly diminished. Supplementary Information The online version contains supplementary material available at 10.1007/s10557-021-07189-9.
Collapse
Affiliation(s)
- Yang Xiao
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Karen Yim
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Hong Zhang
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Diane Bakker
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Rianne Nederlof
- Institut für Herz- und Kreislaufphysiologie, Heinrich- Heine- Universität Düsseldorf, Universitätsstraße 1, Düsseldorf, Germany
| | | | - Herma Renkema
- Khondrion, Philips van Leydenlaan 15, Nijmegen, The Netherlands
| | - Markus W Hollmann
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Nina C Weber
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands
| | - Coert J Zuurbier
- Laboratory of Experimental Intensive Care and Anesthesiology, Department of Anesthesiology, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands.
- Department of Anesthesiology, Amsterdam UMC, Meibergdreef 9, 1105 AZ, Amsterdam, The Netherlands.
| |
Collapse
|
19
|
Prag HA, Gruszczyk AV, Huang MM, Beach TE, Young T, Tronci L, Nikitopoulou E, Mulvey JF, Ascione R, Hadjihambi A, Shattock MJ, Pellerin L, Saeb-Parsy K, Frezza C, James AM, Krieg T, Murphy MP, Aksentijević D. Mechanism of succinate efflux upon reperfusion of the ischaemic heart. Cardiovasc Res 2021; 117:1188-1201. [PMID: 32766828 PMCID: PMC7983001 DOI: 10.1093/cvr/cvaa148] [Citation(s) in RCA: 62] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/13/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
AIMS Succinate accumulates several-fold in the ischaemic heart and is then rapidly oxidized upon reperfusion, contributing to reactive oxygen species production by mitochondria. In addition, a significant amount of the accumulated succinate is released from the heart into the circulation at reperfusion, potentially activating the G-protein-coupled succinate receptor (SUCNR1). However, the factors that determine the proportion of succinate oxidation or release, and the mechanism of this release, are not known. METHODS AND RESULTS To address these questions, we assessed the fate of accumulated succinate upon reperfusion of anoxic cardiomyocytes, and of the ischaemic heart both ex vivo and in vivo. The release of accumulated succinate was selective and was enhanced by acidification of the intracellular milieu. Furthermore, pharmacological inhibition, or haploinsufficiency of the monocarboxylate transporter 1 (MCT1) significantly decreased succinate efflux from the reperfused heart. CONCLUSION Succinate release upon reperfusion of the ischaemic heart is mediated by MCT1 and is facilitated by the acidification of the myocardium during ischaemia. These findings will allow the signalling interaction between succinate released from reperfused ischaemic myocardium and SUCNR1 to be explored.
Collapse
Affiliation(s)
- Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Anja V Gruszczyk
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
- Department of Surgery, University of Cambridge, Cambridge NIHR Biomedical Research Centre, Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Margaret M Huang
- Department of Surgery, University of Cambridge, Cambridge NIHR Biomedical Research Centre, Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Timothy E Beach
- Department of Surgery, University of Cambridge, Cambridge NIHR Biomedical Research Centre, Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Timothy Young
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, PO Box 197, Cambridge CB2 0XZ, UK
| | - Laura Tronci
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, PO Box 197, Cambridge CB2 0XZ, UK
| | - Efterpi Nikitopoulou
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, PO Box 197, Cambridge CB2 0XZ, UK
| | - John F Mulvey
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Raimondo Ascione
- Bristol Medical School and Translational Biomedical Research Centre, Faculty of Health Science, University of Bristol, Level 7, Bristol Royal Infirmary, Upper Maudlin Street, Bristol BS2 8HW, UK
| | - Anna Hadjihambi
- Département de Physiologie, Université de Lausanne, 7 Rue du Bugnon, 1005 Lausanne, Switzerland
| | - Michael J Shattock
- King’s College London, British Heart Foundation Centre of Excellence, The Rayne Institute, St Thomas’ Hospital, Lambeth Palace Road, London SE1 7EH, UK
| | - Luc Pellerin
- Département de Physiologie, Université de Lausanne, 7 Rue du Bugnon, 1005 Lausanne, Switzerland
- Centre de Résonance Magnétique des Systèmes Biologiques, UMR5536 CNRS, LabEx TRAIL-IBIO, Université de Bordeaux, 146 Rue Leo Saignat, Bordeaux 33076, France
- Inserm U1082, Université de Poitiers, 2 Rue de la Miletrie, Poitiers 86021, France
| | - Kourosh Saeb-Parsy
- Department of Surgery, University of Cambridge, Cambridge NIHR Biomedical Research Centre, Biomedical Campus, Hills Road, Cambridge CB2 0QQ, UK
| | - Christian Frezza
- MRC Cancer Unit, University of Cambridge, Hutchison/MRC Research Centre, Cambridge Biomedical Campus, PO Box 197, Cambridge CB2 0XZ, UK
| | - Andrew M James
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0XY, UK
- Department of Medicine, University of Cambridge, Addenbrookes Hospital, Hills Road, Cambridge CB2 0QQ, UK
| | - Dunja Aksentijević
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, Charterhouse Square, London, UK
- Centre for inflammation and Therapeutic Innovation, Queen Mary University of London, Charterhouse Square, London, UK
| |
Collapse
|
20
|
Less Can Be More: The Hormesis Theory of Stress Adaptation in the Global Biosphere and Its Implications. Biomedicines 2021; 9:biomedicines9030293. [PMID: 33805626 PMCID: PMC8000639 DOI: 10.3390/biomedicines9030293] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 03/07/2021] [Accepted: 03/10/2021] [Indexed: 02/07/2023] Open
Abstract
A dose-response relationship to stressors, according to the hormesis theory, is characterized by low-dose stimulation and high-dose inhibition. It is non-linear with a low-dose optimum. Stress responses by cells lead to adapted vitality and fitness. Physical stress can be exerted through heat, radiation, or physical exercise. Chemical stressors include reactive species from oxygen (ROS), nitrogen (RNS), and carbon (RCS), carcinogens, elements, such as lithium (Li) and silicon (Si), and metals, such as silver (Ag), cadmium (Cd), and lead (Pb). Anthropogenic chemicals are agrochemicals (phytotoxins, herbicides), industrial chemicals, and pharmaceuticals. Biochemical stress can be exerted through toxins, medical drugs (e.g., cytostatics, psychopharmaceuticals, non-steroidal inhibitors of inflammation), and through fasting (dietary restriction). Key-lock interactions between enzymes and substrates, antigens and antibodies, antigen-presenting cells, and cognate T cells are the basics of biology, biochemistry, and immunology. Their rules do not obey linear dose-response relationships. The review provides examples of biologic stressors: oncolytic viruses (e.g., immuno-virotherapy of cancer) and hormones (e.g., melatonin, stress hormones). Molecular mechanisms of cellular stress adaptation involve the protein quality control system (PQS) and homeostasis of proteasome, endoplasmic reticulum, and mitochondria. Important components are transcription factors (e.g., Nrf2), micro-RNAs, heat shock proteins, ionic calcium, and enzymes (e.g., glutathion redox enzymes, DNA methyltransferases, and DNA repair enzymes). Cellular growth control, intercellular communication, and resistance to stress from microbial infections involve growth factors, cytokines, chemokines, interferons, and their respective receptors. The effects of hormesis during evolution are multifarious: cell protection and survival, evolutionary flexibility, and epigenetic memory. According to the hormesis theory, this is true for the entire biosphere, e.g., archaia, bacteria, fungi, plants, and the animal kingdoms.
Collapse
|
21
|
Abstract
Under conditions of high nutrient availability and low ATP synthesis, mitochondria generate reactive oxygen species (ROS) that must be removed to avoid cell injury. Among the enzymes involved in this scavenging process, peroxidases play a crucial role, using NADPH provided mostly by nicotinamide nucleotide transhydrogenase (NNT). However, scarce information is available on how and to what extent ROS formation is linked to mitochondrial oxygen consumption. A new study by Smith et al. shows that NNT activity maintains low ROS levels by means of a fine modulation of mitochondrial oxygen utilization.
Collapse
Affiliation(s)
- Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Fabio Di Lisa
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy; Department of Biomedical Sciences, University of Padova, Padova, Italy.
| |
Collapse
|
22
|
Antonucci S, Di Sante M, Tonolo F, Pontarollo L, Scalcon V, Alanova P, Menabò R, Carpi A, Bindoli A, Rigobello MP, Giorgio M, Kaludercic N, Di Lisa F. The Determining Role of Mitochondrial Reactive Oxygen Species Generation and Monoamine Oxidase Activity in Doxorubicin-Induced Cardiotoxicity. Antioxid Redox Signal 2021; 34:531-550. [PMID: 32524823 PMCID: PMC7885901 DOI: 10.1089/ars.2019.7929] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Aims: Doxorubicin cardiomyopathy is a lethal pathology characterized by oxidative stress, mitochondrial dysfunction, and contractile impairment, leading to cell death. Although extensive research has been done to understand the pathophysiology of doxorubicin cardiomyopathy, no effective treatments are available. We investigated whether monoamine oxidases (MAOs) could be involved in doxorubicin-derived oxidative stress, and in the consequent mitochondrial, cardiomyocyte, and cardiac dysfunction. Results: We used neonatal rat ventricular myocytes (NRVMs) and adult mouse ventricular myocytes (AMVMs). Doxorubicin alone (i.e., 0.5 μM doxorubicin) or in combination with H2O2 induced an increase in mitochondrial formation of reactive oxygen species (ROS), which was prevented by the pharmacological inhibition of MAOs in both NRVMs and AMVMs. The pharmacological approach was supported by the genetic ablation of MAO-A in NRVMs. In addition, doxorubicin-derived ROS caused lipid peroxidation and alterations in mitochondrial function (i.e., mitochondrial membrane potential, permeability transition, redox potential), mitochondrial morphology (i.e., mitochondrial distribution and perimeter), sarcomere organization, intracellular [Ca2+] homeostasis, and eventually cell death. All these dysfunctions were abolished by MAO inhibition. Of note, in vivo MAO inhibition prevented chamber dilation and cardiac dysfunction in doxorubicin-treated mice. Innovation and Conclusion: This study demonstrates that the severe oxidative stress induced by doxorubicin requires the involvement of MAOs, which modulate mitochondrial ROS generation. MAO inhibition provides evidence that mitochondrial ROS formation is causally linked to all disorders caused by doxorubicin in vitro and in vivo. Based upon these results, MAO inhibition represents a novel therapeutic approach for doxorubicin cardiomyopathy.
Collapse
Affiliation(s)
| | - Moises Di Sante
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Federica Tonolo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Laura Pontarollo
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Valeria Scalcon
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Petra Alanova
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Institute for Physiology, Czech Academy of Sciences, Prague, Czech Republic
| | - Roberta Menabò
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Andrea Carpi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Alberto Bindoli
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | | | - Marco Giorgio
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,European Institute of Oncology (IEO), Milan, Italy
| | - Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy
| |
Collapse
|
23
|
Yin Z, Burger N, Kula-Alwar D, Aksentijević D, Bridges HR, Prag HA, Grba DN, Viscomi C, James AM, Mottahedin A, Krieg T, Murphy MP, Hirst J. Structural basis for a complex I mutation that blocks pathological ROS production. Nat Commun 2021; 12:707. [PMID: 33514727 PMCID: PMC7846746 DOI: 10.1038/s41467-021-20942-w] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Accepted: 12/23/2020] [Indexed: 02/02/2023] Open
Abstract
Mitochondrial complex I is central to the pathological reactive oxygen species (ROS) production that underlies cardiac ischemia-reperfusion (IR) injury. ND6-P25L mice are homoplasmic for a disease-causing mtDNA point mutation encoding the P25L substitution in the ND6 subunit of complex I. The cryo-EM structure of ND6-P25L complex I revealed subtle structural changes that facilitate rapid conversion to the "deactive" state, usually formed only after prolonged inactivity. Despite its tendency to adopt the "deactive" state, the mutant complex is fully active for NADH oxidation, but cannot generate ROS by reverse electron transfer (RET). ND6-P25L mitochondria function normally, except for their lack of RET ROS production, and ND6-P25L mice are protected against cardiac IR injury in vivo. Thus, this single point mutation in complex I, which does not affect oxidative phosphorylation but renders the complex unable to catalyse RET, demonstrates the pathological role of ROS production by RET during IR injury.
Collapse
Affiliation(s)
- Zhan Yin
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Nils Burger
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | | | - Dunja Aksentijević
- William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London, UK
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University of London, London, UK
| | - Hannah R Bridges
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Daniel N Grba
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Carlo Viscomi
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Department of Biomedical Sciences, University of Padova via Ugo Bassi 58/B, Padova, 35131, Italy
| | - Andrew M James
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Amin Mottahedin
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
- Department of Physiology, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Judy Hirst
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK.
| |
Collapse
|
24
|
Mitochondrial reactive oxygen species in physiology and disease. Cell Calcium 2021; 94:102344. [PMID: 33556741 DOI: 10.1016/j.ceca.2020.102344] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/23/2020] [Accepted: 12/23/2020] [Indexed: 12/13/2022]
Abstract
Mitochondrial reactive oxygen species (mROS) are routinely produced at several sites within the organelle. The balance in their formation and elimination is maintained by a complex and robust antioxidant system. mROS may act as second messengers and regulate a number of physiological processes, such as insulin signaling, cell differentiation and proliferation, wound healing, etc. Nevertheless, when a sudden or sustained increase in ROS formation is not efficiently neutralized by the endogenous antioxidant defense system, the detrimental impact of high mROS levels on cell function and viability eventually results in disease development. In this review, we will focus on the dual role of mROS in pathophysiology, emphasizing the physiological role exerted by a regulated mROS production/elimination, and discussing the detrimental effects evoked by an imbalance in mitochondrial redox state. Furthermore, we will touch upon the interplay between mROS and Ca2+ homeostasis.
Collapse
|
25
|
Cvetko F, Caldwell ST, Higgins M, Suzuki T, Yamamoto M, Prag HA, Hartley RC, Dinkova-Kostova AT, Murphy MP. Nrf2 is activated by disruption of mitochondrial thiol homeostasis but not by enhanced mitochondrial superoxide production. J Biol Chem 2021; 296:100169. [PMID: 33298526 PMCID: PMC7948991 DOI: 10.1074/jbc.ra120.016551] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2020] [Revised: 11/30/2020] [Accepted: 12/09/2020] [Indexed: 12/30/2022] Open
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) regulates the expression of genes involved in antioxidant defenses to modulate fundamental cellular processes such as mitochondrial function and GSH metabolism. Previous reports proposed that mitochondrial reactive oxygen species production and disruption of the GSH pool activate the Nrf2 pathway, suggesting that Nrf2 senses mitochondrial redox signals and/or oxidative damage and signals to the nucleus to respond appropriately. However, until now, it has not been possible to disentangle the overlapping effects of mitochondrial superoxide/hydrogen peroxide production as a redox signal from changes to mitochondrial thiol homeostasis on Nrf2. Recently, we developed mitochondria-targeted reagents that can independently induce mitochondrial superoxide and hydrogen peroxide production mitoParaquat (MitoPQ) or selectively disrupt mitochondrial thiol homeostasis MitoChlorodinitrobenzoic acid (MitoCDNB). Using these reagents, here we have determined how enhanced generation of mitochondrial superoxide and hydrogen peroxide or disruption of mitochondrial thiol homeostasis affects activation of the Nrf2 system in cells, which was assessed by the Nrf2 protein level, nuclear translocation, and expression of its target genes. We found that selective disruption of the mitochondrial GSH pool and inhibition of its thioredoxin system by MitoCDNB led to Nrf2 activation, whereas using MitoPQ to enhance the production of mitochondrial superoxide and hydrogen peroxide alone did not. We further showed that Nrf2 activation by MitoCDNB requires cysteine sensors of Kelch-like ECH-associated protein 1 (Keap1). These findings provide important information on how disruption to mitochondrial redox homeostasis is sensed in the cytoplasm and signaled to the nucleus.
Collapse
Affiliation(s)
- Filip Cvetko
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | | | - Maureen Higgins
- Division of Cellular Medicine, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, Scotland, UK
| | - Takafumi Suzuki
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan; Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Hiran A Prag
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | | | - Albena T Dinkova-Kostova
- Division of Cellular Medicine, School of Medicine, Jacqui Wood Cancer Centre, University of Dundee, Dundee, Scotland, UK; Department of Pharmacology and Molecular Sciences and Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK; Department of Medicine, University of Cambridge, Cambridge, UK.
| |
Collapse
|
26
|
Prag HA, Kula-Alwar D, Pala L, Caldwell ST, Beach TE, James AM, Saeb-Parsy K, Krieg T, Hartley RC, Murphy MP. Selective Delivery of Dicarboxylates to Mitochondria by Conjugation to a Lipophilic Cation via a Cleavable Linker. Mol Pharm 2020; 17:3526-3540. [PMID: 32692564 PMCID: PMC7482397 DOI: 10.1021/acs.molpharmaceut.0c00533] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
![]()
Many
mitochondrial metabolites and bioactive molecules contain
two carboxylic acid moieties that make them unable to cross biological
membranes. Hence, there is considerable interest in facilitating the
uptake of these molecules into cells and mitochondria to modify or
report on their function. Conjugation to the triphenylphosphonium
(TPP) lipophilic cation is widely used to deliver molecules selectively
to mitochondria in response to the membrane potential. However, permanent
attachment to the cation can disrupt the biological function of small
dicarboxylates. Here, we have developed a strategy using TPP to release
dicarboxylates selectively within mitochondria. For this, the dicarboxylate
is attached to a TPP compound via a single ester bond, which is then
cleaved by intramitochondrial esterase activity, releasing the dicarboxylate
within the organelle. Leaving the second carboxylic acid free also
means mitochondrial uptake is dependent on the pH gradient across
the inner membrane. To assess this strategy, we synthesized a range
of TPP monoesters of the model dicarboxylate, malonate. We then tested
their mitochondrial accumulation and ability to deliver malonate to
isolated mitochondria and to cells, in vitro and in vivo. A TPP–malonate monoester compound, TPP11–malonate, in which the dicarboxylate group was attached
to the TPP compound via a hydrophobic undecyl link, was most effective
at releasing malonate within mitochondria in cells and in
vivo. Therefore, we have developed a TPP–monoester
platform that enables the selective release of bioactive dicarboxylates
within mitochondria.
Collapse
Affiliation(s)
- Hiran A Prag
- Molecular Research Center, Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Duvaraka Kula-Alwar
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Laura Pala
- School of Chemistry, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Stuart T Caldwell
- School of Chemistry, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Timothy E Beach
- Department of Surgery, Cambridge National Institute for Health Research Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Andrew M James
- Molecular Research Center, Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge CB2 0XY, United Kingdom
| | - Kourosh Saeb-Parsy
- Department of Surgery, Cambridge National Institute for Health Research Biomedical Research Centre, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| | - Richard C Hartley
- School of Chemistry, University of Glasgow, Glasgow G12 8QQ, United Kingdom
| | - Michael P Murphy
- Molecular Research Center, Mitochondrial Biology Unit, Biomedical Campus, University of Cambridge, Cambridge CB2 0XY, United Kingdom.,Department of Medicine, University of Cambridge, Cambridge CB2 0QQ, United Kingdom
| |
Collapse
|
27
|
Goleva TN, Lyamzaev KG, Rogov AG, Khailova LS, Epremyan KK, Shumakovich GP, Domnina LV, Ivanova OY, Marmiy NV, Zinevich TV, Esipov DS, Zvyagilskaya RA, Skulachev VP, Chernyak BV. Mitochondria-targeted 1,4-naphthoquinone (SkQN) is a powerful prooxidant and cytotoxic agent. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2020; 1861:148210. [PMID: 32305410 DOI: 10.1016/j.bbabio.2020.148210] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/27/2020] [Accepted: 04/14/2020] [Indexed: 12/28/2022]
Abstract
An increase in the production of reactive oxygen species (ROS) in mitochondria due to targeted delivery of redox active compounds may be useful in studies of modulation of cell functions by mitochondrial ROS. Recently, the mitochondria-targeted derivative of menadione (MitoK3) was synthesized. However, MitoK3 did not induce mitochondrial ROS production and lipid peroxidation while exerting significant cytotoxic action. Here we synthesized 1,4-naphthoquinone conjugated with alkyltriphenylphosphonium (SkQN) as a prototype of mitochondria-targeted prooxidant, and its redox properties, interactions with isolated mitochondria, yeast cells and various human cell lines were investigated. According to electrochemical measurements, SkQN was more active redox agent and, due to the absence of methyl group in the naphthoquinone ring, more reactive as electrophile than MitoK3. SkQN (but not MitoK3) stimulated hydrogen peroxide production in isolated mitochondria. At low concentrations, SkQN stimulated state 4 respiration in mitochondria, decreased membrane potential, and blocked ATP synthesis, being more efficient uncoupler of oxidative phosphorylation than MitoK3. In yeast cells, SkQN decreased cell viability and induced oxidative stress and mitochondrial fragmentation. SkQN killed various tumor cells much more efficiently than MitoK3. Since many tumors are characterized by increased oxidative stress, the use of new mitochondria-targeted prooxidants may be a promising strategy for anticancer therapy.
Collapse
Affiliation(s)
- Tatyana N Goleva
- Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Russian Federation
| | - Konstantin G Lyamzaev
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Russian Federation
| | - Anton G Rogov
- Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Russian Federation
| | - Ljudmila S Khailova
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Russian Federation
| | - Khoren K Epremyan
- Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Russian Federation
| | - Galina P Shumakovich
- Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Russian Federation
| | - Lidia V Domnina
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Russian Federation
| | - Olga Yu Ivanova
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Russian Federation
| | - Natalia V Marmiy
- Faculty of Biology, Institute of Mitoengineering, M.V. Lomonosov Moscow State University, Russian Federation
| | - Tatiana V Zinevich
- Faculty of Biology, Institute of Mitoengineering, M.V. Lomonosov Moscow State University, Russian Federation
| | - Dmitry S Esipov
- Faculty of Biology, Institute of Mitoengineering, M.V. Lomonosov Moscow State University, Russian Federation
| | - Renata A Zvyagilskaya
- Bach Institute of Biochemistry, Fundamentals of Biotechnology Federal Research Center, Russian Academy of Sciences, Russian Federation
| | - Vladimir P Skulachev
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Russian Federation
| | - Boris V Chernyak
- A.N. Belozersky Institute of Physico-Chemical Biology, M.V. Lomonosov Moscow State University, Russian Federation.
| |
Collapse
|
28
|
Karoui A, Crochemore C, Harouki N, Corbière C, Preterre D, Vendeville C, Richard V, Fardel O, Lecureur V, Vaugeois JM, Sichel F, Mulder P, Monteil C. Nitrogen Dioxide Inhalation Exposures Induce Cardiac Mitochondrial Reactive Oxygen Species Production, Impair Mitochondrial Function and Promote Coronary Endothelial Dysfunction. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:E5526. [PMID: 32751709 PMCID: PMC7432061 DOI: 10.3390/ijerph17155526] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 07/25/2020] [Accepted: 07/27/2020] [Indexed: 01/01/2023]
Abstract
Traffic air pollution is a major health problem and is recognized as an important risk factor for cardiovascular (CV) diseases. In a previous experimental study, we showed that diesel exhaust (DE) exposures induced cardiac mitochondrial and CV dysfunctions associated with the gaseous phase. Here, we hypothesized that NO2 exposures to levels close to those found in DE induce a mitochondrial reactive oxygen species (ROS) production, which contribute to an endothelial dysfunction, an early indicator for numerous CV diseases. For this, we studied the effects of NO2 on ROS production and its impacts on the mitochondrial, coronary endothelial and cardiac functions, after acute (one single exposure) and repeated (three h/day, five days/week for three weeks) exposures in Wistar rats. Acute NO2 exposure induced an early but reversible mitochondrial ROS production. This event was isolated since neither mitochondrial function nor endothelial function were impaired, whereas cardiac function assessment showed a reversible left ventricular dysfunction. Conversely, after three weeks of exposure this alteration was accompanied by a cardiac mitochondrial dysfunction highlighted by an alteration of adenosine triphosphate (ATP) synthesis and oxidative phosphorylation and an increase in mitochondrial ROS production. Moreover, repeated NO2 exposures promoted endothelial dysfunction of the coronary arteries, as shown by reduced acetylcholine-induced vasodilatation, which was due, at least partially, to a superoxide-dependent decrease of nitric oxide (NO) bioavailability. This study shows that NO2 exposures impair cardiac mitochondrial function, which, in conjunction with coronary endothelial dysfunction, contributes to cardiac dysfunction. Together, these results clearly identify NO2 as a probable risk factor in ischemic heart diseases.
Collapse
Affiliation(s)
- Ahmed Karoui
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 14000 Caen et, 76 000 Rouen, France; (A.K.); (C.C.); (C.C.); (C.V.); (J.-M.V.); (F.S.)
| | - Clément Crochemore
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 14000 Caen et, 76 000 Rouen, France; (A.K.); (C.C.); (C.C.); (C.V.); (J.-M.V.); (F.S.)
| | - Najah Harouki
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, 76 000 Rouen, France; (N.H.); (V.R.); (P.M.)
| | - Cécile Corbière
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 14000 Caen et, 76 000 Rouen, France; (A.K.); (C.C.); (C.C.); (C.V.); (J.-M.V.); (F.S.)
| | - David Preterre
- CERTAM, 1 rue Joseph Fourier, 76 800 Saint-Etienne du Rouvray, France;
| | - Cathy Vendeville
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 14000 Caen et, 76 000 Rouen, France; (A.K.); (C.C.); (C.C.); (C.V.); (J.-M.V.); (F.S.)
| | - Vincent Richard
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, 76 000 Rouen, France; (N.H.); (V.R.); (P.M.)
| | - Olivier Fardel
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)–UMR_S 1085, 35 000 Rennes, France; (O.F.); (V.L.)
- Pôle Biologie, Rennes University Hospital, 35 203 Rennes, France
| | - Valérie Lecureur
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)–UMR_S 1085, 35 000 Rennes, France; (O.F.); (V.L.)
| | - Jean-Marie Vaugeois
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 14000 Caen et, 76 000 Rouen, France; (A.K.); (C.C.); (C.C.); (C.V.); (J.-M.V.); (F.S.)
| | - François Sichel
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 14000 Caen et, 76 000 Rouen, France; (A.K.); (C.C.); (C.C.); (C.V.); (J.-M.V.); (F.S.)
- Centre François Baclesse, 14 000 Caen, France
| | - Paul Mulder
- Normandie Univ, UNIROUEN, Institut National de la Santé et de la Recherche Médicale U1096, 76 000 Rouen, France; (N.H.); (V.R.); (P.M.)
| | - Christelle Monteil
- Normandie Univ, UNIROUEN, UNICAEN ABTE, 14000 Caen et, 76 000 Rouen, France; (A.K.); (C.C.); (C.C.); (C.V.); (J.-M.V.); (F.S.)
| |
Collapse
|
29
|
Chowdhury AR, Zielonka J, Kalyanaraman B, Hartley RC, Murphy MP, Avadhani NG. Mitochondria-targeted paraquat and metformin mediate ROS production to induce multiple pathways of retrograde signaling: A dose-dependent phenomenon. Redox Biol 2020; 36:101606. [PMID: 32604037 PMCID: PMC7327929 DOI: 10.1016/j.redox.2020.101606] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Accepted: 06/11/2020] [Indexed: 01/12/2023] Open
Abstract
The mitochondrial electron transport chain is a major source of reactive oxygen species (ROS) and is also a target of ROS, with an implied role in the stabilization of hypoxia-inducible factor (HIF) and induction of the AMPK pathway. Here we used varying doses of two agents, Mito-Paraquat and Mito-Metformin, that have been conjugated to cationic triphenylphosphonium (TPP+) moiety to selectively target them to the mitochondrial matrix compartment, thereby resulting in the site-specific generation of ROS within mitochondria. These agents primarily induce superoxide (O2•-) production by acting on complex I. In Raw264.7 macrophages, C2C12 skeletal myocytes, and HCT116 adenocarcinoma cells, we show that mitochondria-targeted oxidants can induce ROS (O2•- and H2O2). In all three cell lines tested, the mitochondria-targeted agents disrupted membrane potential and activated calcineurin and the Cn-dependent retrograde signaling pathway. Hypoxic culture conditions also induced Cn activation and HIF1α activation in a temporally regulated manner, with the former appearing at shorter exposure times. Together, our results indicate that mitochondrial oxidant-induced retrograde signaling is driven by disruption of membrane potential and activation of Ca2+/Cn pathway and is independent of ROS-induced HIF1α or AMPK pathways.
Collapse
Affiliation(s)
- Anindya Roy Chowdhury
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacek Zielonka
- Department of Biophysics and Free Radical Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Balaraman Kalyanaraman
- Department of Biophysics and Free Radical Research Center, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Michael P Murphy
- MRC-Mitochondrial Biology Unit, University of Cambridge, Hills Road, Cambridge, CB2 OXY, UK
| | - Narayan G Avadhani
- Department of Biomedical Sciences, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
30
|
Andreadou I, Schulz R, Papapetropoulos A, Turan B, Ytrehus K, Ferdinandy P, Daiber A, Di Lisa F. The role of mitochondrial reactive oxygen species, NO and H 2 S in ischaemia/reperfusion injury and cardioprotection. J Cell Mol Med 2020; 24:6510-6522. [PMID: 32383522 PMCID: PMC7299678 DOI: 10.1111/jcmm.15279] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2020] [Revised: 03/04/2020] [Accepted: 03/08/2020] [Indexed: 12/12/2022] Open
Abstract
Redox signalling in mitochondria plays an important role in myocardial ischaemia/reperfusion (I/R) injury and in cardioprotection. Reactive oxygen and nitrogen species (ROS/RNS) modify cellular structures and functions by means of covalent changes in proteins including among others S‐nitros(yl)ation by nitric oxide (NO) and its derivatives, and S‐sulphydration by hydrogen sulphide (H2S). Many enzymes are involved in the mitochondrial formation and handling of ROS, NO and H2S under physiological and pathological conditions. In particular, the balance between formation and removal of reactive species is impaired during I/R favouring their accumulation. Therefore, various interventions aimed at decreasing mitochondrial ROS accumulation have been developed and have shown cardioprotective effects in experimental settings. However, ROS, NO and H2S play also a role in endogenous cardioprotection, as in the case of ischaemic pre‐conditioning, so that preventing their increase might hamper self‐defence mechanisms. The aim of the present review was to provide a critical analysis of formation and role of reactive species, NO and H2S in mitochondria, with a special emphasis on mechanisms of injury and protection that determine the fate of hearts subjected to I/R. The elucidation of the signalling pathways of ROS, NO and H2S is likely to reveal novel molecular targets for cardioprotection that could be modulated by pharmacological agents to prevent I/R injury.
Collapse
Affiliation(s)
- Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig University Giessen, Giessen, Germany
| | - Andreas Papapetropoulos
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Belma Turan
- Department of Biophysics, Faculty of Medicine, Ankara University, Ankara, Turkey
| | - Kirsti Ytrehus
- Department of Medical Biology, UiT The Arctic University of Norway, Tromso, Norway
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.,Pharmahungary Group, Szeged, Hungary
| | - Andreas Daiber
- Molecular Cardiology, Center for Cardiology 1, University Medical Center of the Johannes Gutenberg University, Mainz, Germany
| | - Fabio Di Lisa
- Department of Biomedical Sciences, Università degli Studi di Padova, Padova, Italy
| |
Collapse
|
31
|
Li B, Zhao H, Wu Y, Zhu Y, Zhang J, Yang G, Yan Q, Li J, Li T, Liu L. Mitochondrial-Derived Vesicles Protect Cardiomyocytes Against Hypoxic Damage. Front Cell Dev Biol 2020; 8:214. [PMID: 32426351 PMCID: PMC7212461 DOI: 10.3389/fcell.2020.00214] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 03/11/2020] [Indexed: 01/20/2023] Open
Abstract
Myocardial ischemia is a condition with insufficient oxygen supporting the heart tissues, which may result from myocardial infarction or trauma-induced hemorrhagic shock. In order to develop better preventive and therapeutic strategies for myocardial ischemic damage, it is important that we understand the mechanisms underlying this type of injury. Mitochondrial-derived vesicles (MDVs) have been proposed as a novel player in maintaining mitochondrial quality control. This study aimed to investigate the role and possible mechanisms of MDVs in ischemia/hypoxia-induced myocardial apoptosis. H9C2 cardiomyocytes were used for the cellular experiments. A 40% fixed blood volume hemorrhagic shock rat model was used to construct the acute general ischemic models. MDVs were detected using immunofluorescence staining with PDH and TOM20. Exogenous MDVs were reconstituted in vitro from isolated mitochondria under different hypoxic conditions. The results demonstrate that MDV production was negatively correlated with cardiomyocyte apoptosis under hypoxic conditions; exogenous MDVs inhibited hypoxia-induced cardiomyocyte apoptosis; and MDV-mediated protection against hypoxia-induced cardiomyocyte apoptosis was accomplished via Bcl-2 interactions in the mitochondrial pathway. This study provides evidence that MDVs protect cardiomyocytes against hypoxic damage by inhibiting mitochondrial apoptosis. Our study used a novel approach that expands our understanding of MDVs and highlights that MDVs may be part of the endogenous response to hypoxia designed to mitigate damage. Strategies that stimulate cardiomyocytes to produce cargo-specific MDVs, including Bcl-2 containing MDVs, could theoretically be helpful in treating ischemic/hypoxic myocardial injury.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Tao Li
- State Key Laboratory of Trauma, Burns and Combined Injury, Department 2, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| | - Liangming Liu
- State Key Laboratory of Trauma, Burns and Combined Injury, Department 2, Research Institute of Surgery, Daping Hospital, Army Medical University, Chongqing, China
| |
Collapse
|
32
|
Szibor M, Schreckenberg R, Gizatullina Z, Dufour E, Wiesnet M, Dhandapani PK, Debska‐Vielhaber G, Heidler J, Wittig I, Nyman TA, Gärtner U, Hall AR, Pell V, Viscomi C, Krieg T, Murphy MP, Braun T, Gellerich FN, Schlüter K, Jacobs HT. Respiratory chain signalling is essential for adaptive remodelling following cardiac ischaemia. J Cell Mol Med 2020; 24:3534-3548. [PMID: 32040259 PMCID: PMC7131948 DOI: 10.1111/jcmm.15043] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Revised: 01/19/2020] [Accepted: 01/21/2020] [Indexed: 01/09/2023] Open
Abstract
Cardiac ischaemia-reperfusion (I/R) injury has been attributed to stress signals arising from an impaired mitochondrial electron transport chain (ETC), which include redox imbalance, metabolic stalling and excessive production of reactive oxygen species (ROS). The alternative oxidase (AOX) is a respiratory enzyme, absent in mammals, that accepts electrons from a reduced quinone pool to reduce oxygen to water, thereby restoring electron flux when impaired and, in the process, blunting ROS production. Hence, AOX represents a natural rescue mechanism from respiratory stress. This study aimed to determine how respiratory restoration through xenotopically expressed AOX affects the re-perfused post-ischaemic mouse heart. As expected, AOX supports ETC function and attenuates the ROS load in post-anoxic heart mitochondria. However, post-ischaemic cardiac remodelling over 3 and 9 weeks was not improved. AOX blunted transcript levels of factors known to be up-regulated upon I/R such as the atrial natriuretic peptide (Anp) whilst expression of pro-fibrotic and pro-apoptotic transcripts were increased. Ex vivo analysis revealed contractile failure at nine but not 3 weeks after ischaemia whilst label-free quantitative proteomics identified an increase in proteins promoting adverse extracellular matrix remodelling. Together, this indicates an essential role for ETC-derived signals during cardiac adaptive remodelling and identified ROS as a possible effector.
Collapse
Affiliation(s)
- Marten Szibor
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
- Department of Cardiothoracic SurgeryJena University HospitalJenaGermany
| | | | | | - Eric Dufour
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
| | - Marion Wiesnet
- Department Cardiac Development and RemodellingMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | - Praveen K. Dhandapani
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
| | | | - Juliana Heidler
- Functional ProteomicsFaculty of MedicineGoethe UniversityFrankfurt am MainGermany
| | - Ilka Wittig
- Functional ProteomicsFaculty of MedicineGoethe UniversityFrankfurt am MainGermany
| | - Tuula A. Nyman
- Department of ImmunologyInstitute of Clinical MedicineOslo University HospitalUniversity of OsloOsloNorway
| | - Ulrich Gärtner
- Institute of Anatomy and Cell BiologyJustus‐Liebig‐University GiessenGiessenGermany
| | - Andrew R. Hall
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUK
| | - Victoria Pell
- Department of MedicineUniversity of CambridgeCambridgeUK
| | - Carlo Viscomi
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUK
- Department of Biomedical SciencesUniversity of PadovaPadovaItaly
| | - Thomas Krieg
- Department of MedicineUniversity of CambridgeCambridgeUK
| | - Michael P. Murphy
- Medical Research Council Mitochondrial Biology UnitUniversity of CambridgeCambridgeUK
| | - Thomas Braun
- Department Cardiac Development and RemodellingMax Planck Institute for Heart and Lung ResearchBad NauheimGermany
| | | | | | - Howard T. Jacobs
- Faculty of Medicine and Health TechnologyTampere UniversityTampereFinland
- Institute of BiotechnologyUniversity of HelsinkiHelsinkiFinland
| |
Collapse
|
33
|
Kaludercic N, Di Lisa F. Mitochondrial ROS Formation in the Pathogenesis of Diabetic Cardiomyopathy. Front Cardiovasc Med 2020; 7:12. [PMID: 32133373 PMCID: PMC7040199 DOI: 10.3389/fcvm.2020.00012] [Citation(s) in RCA: 162] [Impact Index Per Article: 32.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Accepted: 01/28/2020] [Indexed: 12/20/2022] Open
Abstract
Diabetic cardiomyopathy is a result of diabetes-induced changes in the structure and function of the heart. Hyperglycemia affects multiple pathways in the diabetic heart, but excessive reactive oxygen species (ROS) generation and oxidative stress represent common denominators associated with adverse tissue remodeling. Indeed, key processes underlying cardiac remodeling in diabetes are redox sensitive, including inflammation, organelle dysfunction, alteration in ion homeostasis, cardiomyocyte hypertrophy, apoptosis, fibrosis, and contractile dysfunction. Extensive experimental evidence supports the involvement of mitochondrial ROS formation in the alterations characterizing the diabetic heart. In this review we will outline the central role of mitochondrial ROS and alterations in the redox status contributing to the development of diabetic cardiomyopathy. We will discuss the role of different sources of ROS involved in this process, with a specific emphasis on mitochondrial ROS producing enzymes within cardiomyocytes. Finally, the therapeutic potential of pharmacological inhibitors of ROS sources within the mitochondria will be discussed.
Collapse
Affiliation(s)
- Nina Kaludercic
- Neuroscience Institute, National Research Council of Italy (CNR), Padua, Italy
| | - Fabio Di Lisa
- Neuroscience Institute, National Research Council of Italy (CNR), Padua, Italy.,Department of Biomedical Sciences, University of Padua, Padua, Italy
| |
Collapse
|
34
|
Rhooms SK, Murari A, Goparaju NSV, Vilanueva M, Owusu-Ansah E. Insights from Drosophila on mitochondrial complex I. Cell Mol Life Sci 2020; 77:607-618. [PMID: 31485716 PMCID: PMC7289077 DOI: 10.1007/s00018-019-03293-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 08/08/2019] [Accepted: 08/28/2019] [Indexed: 12/26/2022]
Abstract
NADH:ubiquinone oxidoreductase, more commonly referred to as mitochondrial complex I (CI), is the largest discrete enzyme of the oxidative phosphorylation system (OXPHOS). It is localized to the mitochondrial inner membrane. CI oxidizes NADH generated from the tricarboxylic acid cycle to NAD+, in a series of redox reactions that culminates in the reduction of ubiquinone, and the transport of protons from the matrix across the inner membrane to the intermembrane space. The resulting proton-motive force is consumed by ATP synthase to generate ATP, or harnessed to transport ions, metabolites and proteins into the mitochondrion. CI is also a major source of reactive oxygen species. Accordingly, impaired CI function has been associated with a host of chronic metabolic and degenerative disorders such as diabetes, cardiomyopathy, Parkinson's disease (PD) and Leigh syndrome. Studies on Drosophila have contributed to our understanding of the multiple roles of CI in bioenergetics and organismal physiology. Here, we explore and discuss some of the studies on Drosophila that have informed our understanding of this complex and conclude with some of the open questions about CI that can be resolved by studies on Drosophila.
Collapse
Affiliation(s)
- Shauna-Kay Rhooms
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, 10032, USA
| | - Anjaneyulu Murari
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, 10032, USA
| | - Naga Sri Vidya Goparaju
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, 10032, USA
| | - Maximino Vilanueva
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, 10032, USA
| | - Edward Owusu-Ansah
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY, 10032, USA.
- The Robert N. Butler Columbia Aging Center, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|