9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      The role of mitochondrial reactive oxygen species, NO and H 2S in ischaemia/reperfusion injury and cardioprotection

      review-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          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 (H 2S). Many enzymes are involved in the mitochondrial formation and handling of ROS, NO and H 2S 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 H 2S 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 H 2S 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 H 2S is likely to reveal novel molecular targets for cardioprotection that could be modulated by pharmacological agents to prevent I/R injury.

          Related collections

          Most cited references120

          • Record: found
          • Abstract: found
          • Article: not found

          The sites and topology of mitochondrial superoxide production.

          Mitochondrial superoxide production is an important source of reactive oxygen species in cells, and may cause or contribute to ageing and the diseases of ageing. Seven major sites of superoxide production in mammalian mitochondria are known and widely accepted. In descending order of maximum capacity they are the ubiquinone-binding sites in complex I (site IQ) and complex III (site IIIQo), glycerol 3-phosphate dehydrogenase, the flavin in complex I (site IF), the electron transferring flavoprotein:Q oxidoreductase (ETFQOR) of fatty acid beta-oxidation, and pyruvate and 2-oxoglutarate dehydrogenases. None of these sites is fully characterized and for some we only have sketchy information. The topology of the sites is important because it determines whether or not a site will produce superoxide in the mitochondrial matrix and be able to damage mitochondrial DNA. All sites produce superoxide in the matrix; site IIIQo and glycerol 3-phosphate dehydrogenase also produce superoxide to the intermembrane space. The relative contribution of each site to mitochondrial reactive oxygen species generation in the absence of electron transport inhibitors is unknown in isolated mitochondria, in cells or in vivo, and may vary considerably with species, tissue, substrate, energy demand and oxygen tension. Copyright (c) 2010 Elsevier Inc. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Dilated cardiomyopathy and neonatal lethality in mutant mice lacking manganese superoxide dismutase.

            The Sod2 gene for Mn-superoxide dismutase (MnSOD), an intramitochondrial free radical scavenging enzyme that is the first line of defense against superoxide produced as a byproduct of oxidative phosphorylation, was inactivated by homologous recombination. Homozygous mutant mice die within the first 10 days of life with a dilated cardiomyopathy, accumulation of lipid in liver and skeletal muscle, and metabolic acidosis. Cytochemical analysis revealed a severe reduction in succinate dehydrogenase (complex II) and aconitase (a TCA cycle enzyme) activities in the heart and, to a lesser extent, in other organs. These findings indicate that MnSOD is required for normal biological function of tissues by maintaining the integrity of mitochondrial enzymes susceptible to direct inactivation by superoxide.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Mitohormesis.

              For many years, mitochondria were viewed as semiautonomous organelles, required only for cellular energetics. This view has been largely supplanted by the concept that mitochondria are fully integrated into the cell and that mitochondrial stresses rapidly activate cytosolic signaling pathways that ultimately alter nuclear gene expression. Remarkably, this coordinated response to mild mitochondrial stress appears to leave the cell less susceptible to subsequent perturbations. This response, termed mitohormesis, is being rapidly dissected in many model organisms. A fuller understanding of mitohormesis promises to provide insight into our susceptibility for disease and potentially provide a unifying hypothesis for why we age. Copyright © 2014 Elsevier Inc. All rights reserved.
                Bookmark

                Author and article information

                Contributors
                daiber@uni-mainz.de
                dilisa@bio.unipd.it
                Journal
                J Cell Mol Med
                J. Cell. Mol. Med
                10.1111/(ISSN)1582-4934
                JCMM
                Journal of Cellular and Molecular Medicine
                John Wiley and Sons Inc. (Hoboken )
                1582-1838
                1582-4934
                08 May 2020
                June 2020
                : 24
                : 12 ( doiID: 10.1111/jcmm.v24.12 )
                : 6510-6522
                Affiliations
                [ 1 ] Laboratory of Pharmacology Faculty of Pharmacy National and Kapodistrian University of Athens Athens Greece
                [ 2 ] Institute for Physiology Justus‐Liebig University Giessen Giessen Germany
                [ 3 ] Department of Biophysics Faculty of Medicine Ankara University Ankara Turkey
                [ 4 ] Department of Medical Biology UiT The Arctic University of Norway Tromso Norway
                [ 5 ] Department of Pharmacology and Pharmacotherapy Semmelweis University Budapest Hungary
                [ 6 ] Pharmahungary Group Szeged Hungary
                [ 7 ] Molecular Cardiology Center for Cardiology 1 University Medical Center of the Johannes Gutenberg University Mainz Germany
                [ 8 ] Department of Biomedical Sciences Università degli Studi di Padova Padova Italy
                Author notes
                [*] [* ] Correspondence

                Fabio Di Lisa, Department of Biomedical Sciences, Università degli Studi di Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy.

                Email: dilisa@ 123456bio.unipd.it

                and

                Andreas Daiber, Universitätsmedizin Mainz, Zentrum für Kardiologie 1, Labor für Molekulare Kardiologie, Langenbeckstr. 1, 55131 Mainz, Germany.

                Email: daiber@ 123456uni-mainz.de

                Author information
                https://orcid.org/0000-0003-2583-9294
                https://orcid.org/0000-0002-2769-0094
                https://orcid.org/0000-0003-0705-7539
                Article
                JCMM15279
                10.1111/jcmm.15279
                7299678
                32383522
                3b4667b9-5c38-45d8-bd64-36478463095e
                © 2020 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 08 January 2020
                : 04 March 2020
                : 08 March 2020
                Page count
                Figures: 2, Tables: 0, Pages: 13, Words: 11258
                Funding
                Funded by: National Research, Development and Innovation Office of Hungary
                Award ID: NVKP 16‐1‐2016‐0017
                Funded by: Hellenic Foundation for Research and Innovation
                Award ID: HFRI‐FM17‐886
                Funded by: European Regional Development Fund , open-funder-registry 10.13039/501100008530;
                Award ID: 5002803
                Funded by: Fondation Leducq , open-funder-registry 10.13039/501100001674;
                Award ID: 16CVD04
                Funded by: European Cooperation in Science and Technology , open-funder-registry 10.13039/501100000921;
                Award ID: CA16225
                Funded by: Higher Education Institutional Excellence Program of the Ministry of Human Capacities in Hungary
                Categories
                Review
                Reviews
                Custom metadata
                2.0
                June 2020
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.8.4 mode:remove_FC converted:17.06.2020

                Molecular medicine
                cardioprotection,heart,hydrogen sulphide,ischaemia,mitochondria,nitric oxide,reactive oxygen species,reperfusion

                Comments

                Comment on this article