1
|
Mazidi Z, Wieser M, Spinu N, Weidinger A, Kozlov AV, Vukovic K, Wellens S, Murphy C, Singh P, Lagares LM, Bobbili MR, Liendl L, Schosserer M, Diendorfer A, Bettelheim B, Eilenberg W, Exner T, Culot M, Jennings P, Wilmes A, Novic M, Benfenati E, Grillari-Voglauer R, Grillari J. Cyclosporin A toxicity on endothelial cells differentiated from induced pluripotent stem cells: Assembling an adverse outcome pathway. Toxicol In Vitro 2025; 103:105954. [PMID: 39550010 DOI: 10.1016/j.tiv.2024.105954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 06/15/2024] [Accepted: 10/15/2024] [Indexed: 11/18/2024]
Abstract
Cyclosporin A (CSA) is a potent immunosuppressive agent in pharmacologic studies. However, there is evidence for side effects, specifically regarding vascular dysfunction. Its mode of action inducing endothelial cell toxicity is partially unclear, and a connection with an adverse outcome pathway (AOP) is not established yet. Therefore, we designed this study to get deeper insights into the mechanistic toxicology of CSA on angiogenesis. Stem cells, especially induced pluripotent stem cells (iPSCs) with the ability of differentiation to all organs of the body, are considered a promising in vitro model to reduce animal experimentation. In this study, we differentiated iPSCs to endothelial cells (ECs) as one cell type that in other studies would allow to generate multi-cell type organoids from single donors. Flow cytometry and immunostaining confirmed our scalable differentiation protocol. Then dose and time course experiments assessing CSA cytotoxicity on iPS derived endothelial cells were performed. Transcriptomic data suggested CSA dependent induction of reactive oxygen species (ROS), mitochondrial dysfunction, and impaired angiogenesis via ROS induction which was confirmed by in vitro experiments. In order to put these data into a potential adverse outcome pathway (AOP) context, we performed a literature review for CSA-mediated endothelial cell toxicity and combined our experimental data with the publicly available knowledge. Such an AOP will help to design in vitro test batteries and to model events observed in human toxicity studies, as well in predictive toxicology.
Collapse
Affiliation(s)
- Zahra Mazidi
- Evercyte GmbH, Leberstrasse 20, 1110 Vienna, Austria; Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Muthgasse 18, 1190 Vienna, Austria
| | | | - Nicoleta Spinu
- School of Pharmacy and Biomolecular Sciences, Liverpool John Moores University, Byrom Street, Liverpool L3 3AF, UK
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstrasse 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Andrey V Kozlov
- Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstrasse 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Kristijan Vukovic
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche "Mario Negri"-IRCCS, Via Mario Negri 2, 20156 Milano, Italy
| | - Sara Wellens
- University of Artois, UR2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, Rue Jean Souvraz SP18, F-62300 Lens, France
| | - Cormac Murphy
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081HZ Amsterdam, the Netherlands
| | - Pranika Singh
- Edelweiss Connect GmbH, Technology Park Basel, Hochbergerstrasse 60C, 4057 Basel, Switzerland; Division of Molecular and Systems Toxicology, Department of Pharmaceutical Sciences, University of Basel, Klingelbergstrasse 50, 4056 Basel, Switzerland
| | - Liadys Mora Lagares
- Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Madhusudhan Reddy Bobbili
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Muthgasse 18, 1190 Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstrasse 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Lisa Liendl
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Muthgasse 18, 1190 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Markus Schosserer
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Muthgasse 18, 1190 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | | | - Bruno Bettelheim
- Department of Obstetrics and Gynecology, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Wolf Eilenberg
- Department of General Surgery, Medical University of Vienna, Währinger Gürtel 18-20, 1090 Vienna, Austria
| | - Thomas Exner
- Seven Past Nine d.o.o., Hribljane 10, 1380 Cerknica, Slovenia
| | - Maxime Culot
- University of Artois, UR2465, Laboratoire de la Barrière Hémato-Encéphalique (LBHE), Faculté des sciences Jean Perrin, Rue Jean Souvraz SP18, F-62300 Lens, France
| | - Paul Jennings
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081HZ Amsterdam, the Netherlands
| | - Anja Wilmes
- Division of Molecular and Computational Toxicology, Department of Chemistry and Pharmaceutical Sciences, Amsterdam Institute for Molecules, Medicines and Systems, Vrije Universiteit Amsterdam, De Boelelaan 1108, 1081HZ Amsterdam, the Netherlands
| | - Marjana Novic
- Theory Department, Laboratory for Cheminformatics, National Institute of Chemistry, 1000 Ljubljana, Slovenia
| | - Emilio Benfenati
- Laboratory of Environmental Chemistry and Toxicology, Department of Environmental Health Sciences, Istituto di Ricerche Farmacologiche "Mario Negri"-IRCCS, Via Mario Negri 2, 20156 Milano, Italy
| | - Regina Grillari-Voglauer
- Evercyte GmbH, Leberstrasse 20, 1110 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Johannes Grillari
- Institute of Molecular Biotechnology, Department of Biotechnology, BOKU University, Muthgasse 18, 1190 Vienna, Austria; Ludwig Boltzmann Institute for Traumatology, The Research Center in Cooperation with AUVA, Donaueschingenstrasse 13, 1200 Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria.
| |
Collapse
|
2
|
Tichauer JE, Rovegno M. Role of astrocytes connexins - pannexins in acute brain injury. Neurotherapeutics 2025; 22:e00523. [PMID: 39848901 PMCID: PMC11840357 DOI: 10.1016/j.neurot.2025.e00523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/25/2025] Open
Abstract
Acute brain injuries (ABIs) encompass a broad spectrum of primary injuries such as ischemia, hypoxia, trauma, and hemorrhage that converge into secondary injury where some mechanisms show common determinants. In this regard, astroglial connexin and pannexin channels have been shown to play an important role. These channels are transmembrane proteins sharing similar topology and form gateways between adjacent cells named gap junctions (GJs) and pores into unopposed membranes named hemichannels (HCs). In astrocytes, GJs and HCs enable intercellular communication and have active participation in normal brain physiological processes, such as calcium waves, synapsis modulation, regional blood flow regulation, and homeostatic control of the extracellular environment, among others. However, after acute brain injury, astrocytes can change their phenotype and modify the activity of both channels and hemichannels, which can result in the amplification of danger signals, increased mediators of inflammation, and neuronal death, contributing to the expansion of brain damage and neurological deterioration. This is known as secondary brain damage. In this review, we discussed the main biological mechanism of secondary brain damage with a particular focus on astroglial connexin and pannexin participation during acute brain injuries.
Collapse
Affiliation(s)
- Juan E Tichauer
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile.
| | - Maximiliano Rovegno
- Departamento de Medicina Intensiva, Facultad de Medicina, Pontificia Universidad Católica de Chile, Chile.
| |
Collapse
|
3
|
Gerle C, Jiko C, Nakano A, Yokoyama K, Gopalasingam CC, Shigematsu H, Abe K. Human F-ATP synthase as a drug target. Pharmacol Res 2024; 209:107423. [PMID: 39303772 DOI: 10.1016/j.phrs.2024.107423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
Practical and conceptual barriers have kept human F-ATP synthase out of reach as a target for the treatment of human diseases. Although this situation has persisted for decades, it may change in the near future. In this review the principal functionalities of human F-ATP synthase--proton motive force / ATP interconversion, membrane bending and mitochondrial permeability transition--are surveyed in the context of their respective potential for pharmaceutical intervention. Further, the technical requirements necessary to allow drug designs that are effective at the multiple levels of functionality and modality of human F-ATP synthase are discussed. The structure-based development of gastric proton pump inhibitors is used to exemplify what might be feasible for human F-ATP synthase. And finally, four structural regions of the human F-ATP synthase are examined as potential sites for the development of structure based drug development.
Collapse
Affiliation(s)
- Christoph Gerle
- Life Science Research Infrastructure Group, RIKEN SPring-8 Center, Kouto, 1-1-1, Sayo, Hyogo, Japan.
| | - Chimari Jiko
- Division of Radiation Life Science, Institute for Integrated Radiation and Nuclear Science, Kyoto University, Osaka, Japan
| | - Atsuki Nakano
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kyoto 603-8555, Japan
| | - Ken Yokoyama
- Department of Molecular Biosciences, Kyoto Sangyo University, Kamigamo-Motoyama, Kyoto 603-8555, Japan
| | - Chai C Gopalasingam
- Life Science Research Infrastructure Group, RIKEN SPring-8 Center, Kouto, 1-1-1, Sayo, Hyogo, Japan
| | - Hideki Shigematsu
- Structural Biology Division, Japan Synchrotron Radiation Research Institute, SPring-8, Sayo, Hyogo, Japan
| | - Kazuhiro Abe
- Molecular Biochemistry Lab, Department of Chemistry, Faculty of Science, Hokkaido University, Sapporo 060-0810, Japan
| |
Collapse
|
4
|
Zorova LD, Abramicheva PA, Andrianova NV, Babenko VA, Zorov SD, Pevzner IB, Popkov VA, Semenovich DS, Yakupova EI, Silachev DN, Plotnikov EY, Sukhikh GT, Zorov DB. Targeting Mitochondria for Cancer Treatment. Pharmaceutics 2024; 16:444. [PMID: 38675106 PMCID: PMC11054825 DOI: 10.3390/pharmaceutics16040444] [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: 02/28/2024] [Accepted: 03/20/2024] [Indexed: 04/28/2024] Open
Abstract
There is an increasing accumulation of data on the exceptional importance of mitochondria in the occurrence and treatment of cancer, and in all lines of evidence for such participation, there are both energetic and non-bioenergetic functional features of mitochondria. This analytical review examines three specific features of adaptive mitochondrial changes in several malignant tumors. The first feature is characteristic of solid tumors, whose cells are forced to rebuild their energetics due to the absence of oxygen, namely, to activate the fumarate reductase pathway instead of the traditional succinate oxidase pathway that exists in aerobic conditions. For such a restructuring, the presence of a low-potential quinone is necessary, which cannot ensure the conventional conversion of succinate into fumarate but rather enables the reverse reaction, that is, the conversion of fumarate into succinate. In this scenario, complex I becomes the only generator of energy in mitochondria. The second feature is the increased proliferation in aggressive tumors of the so-called mitochondrial (peripheral) benzodiazepine receptor, also called translocator protein (TSPO) residing in the outer mitochondrial membrane, the function of which in oncogenic transformation stays mysterious. The third feature of tumor cells is the enhanced retention of certain molecules, in particular mitochondrially directed cations similar to rhodamine 123, which allows for the selective accumulation of anticancer drugs in mitochondria. These three features of mitochondria can be targets for the development of an anti-cancer strategy.
Collapse
Affiliation(s)
- Ljubava D. Zorova
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Polina A. Abramicheva
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
| | - Nadezda V. Andrianova
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
| | - Valentina A. Babenko
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Savva D. Zorov
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Irina B. Pevzner
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Vasily A. Popkov
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Dmitry S. Semenovich
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
| | - Elmira I. Yakupova
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
| | - Denis N. Silachev
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
| | - Egor Y. Plotnikov
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Gennady T. Sukhikh
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| | - Dmitry B. Zorov
- A.N. Belozersky Research Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia; (L.D.Z.); (P.A.A.); (V.A.B.); (S.D.Z.); (I.B.P.); (V.A.P.); (D.S.S.); (E.I.Y.); (D.N.S.); (E.Y.P.)
- V.I. Kulakov National Medical Research Center of Obstetrics, Gynecology and Perinatology, 117997 Moscow, Russia
| |
Collapse
|
5
|
Neginskaya MA, Pavlov EV. Investigation of Properties of the Mitochondrial Permeability Transition Pore Using Whole-Mitoplast Patch-Clamp Technique. DNA Cell Biol 2023; 42:481-487. [PMID: 37311169 PMCID: PMC10460960 DOI: 10.1089/dna.2023.0171] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 06/15/2023] Open
Abstract
The mitochondrial permeability transition pore (mPTP) is a channel in the mitochondrial inner membrane that is activated by excessive calcium uptake. In this study, we used a whole-mitoplast patch-clamp approach to investigate the ionic currents associated with mPTP at the level of the whole single mitochondrion. The whole-mitoplast conductance was at the level of 5 to 7 nS, which is consistent with the presence of three to six single mPTP channels per mitochondrion. We found that mPTP currents are voltage dependent and inactivate at negative potential. The currents were inhibited by cyclosporine A and adenosine diphosphate. When mPTP was induced by oxidative stress, currents were partially blocked by the adenine nucleotide translocase inhibitor bongkrekic acid. Our data suggest that the whole-mitoplast patch-clamp approach is a useful method for investigating the biophysical properties and regulation of the mPTP.
Collapse
Affiliation(s)
- Maria A. Neginskaya
- Department of Molecular Pathobiology, New York University, New York, New York, USA
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Evgeny V. Pavlov
- Department of Molecular Pathobiology, New York University, New York, New York, USA
| |
Collapse
|
6
|
Chapa-Dubocq XR, Rodríguez-Graciani KM, Escobales N, Javadov S. Mitochondrial Volume Regulation and Swelling Mechanisms in Cardiomyocytes. Antioxidants (Basel) 2023; 12:1517. [PMID: 37627512 PMCID: PMC10451443 DOI: 10.3390/antiox12081517] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Mitochondrion, known as the "powerhouse" of the cell, regulates ion homeostasis, redox state, cell proliferation and differentiation, and lipid synthesis. The inner mitochondrial membrane (IMM) controls mitochondrial metabolism and function. It possesses high levels of proteins that account for ~70% of the membrane mass and are involved in the electron transport chain, oxidative phosphorylation, energy transfer, and ion transport, among others. The mitochondrial matrix volume plays a crucial role in IMM remodeling. Several ion transport mechanisms, particularly K+ and Ca2+, regulate matrix volume. Small increases in matrix volume through IMM alterations can activate mitochondrial respiration, whereas excessive swelling can impair the IMM topology and initiates mitochondria-mediated cell death. The opening of mitochondrial permeability transition pores, the well-characterized phenomenon with unknown molecular identity, in low- and high-conductance modes are involved in physiological and pathological increases of matrix volume. Despite extensive studies, the precise mechanisms underlying changes in matrix volume and IMM structural remodeling in response to energy and oxidative stressors remain unknown. This review summarizes and discusses previous studies on the mechanisms involved in regulating mitochondrial matrix volume, IMM remodeling, and the crosstalk between these processes.
Collapse
Affiliation(s)
| | | | | | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA; (X.R.C.-D.); (K.M.R.-G.); (N.E.)
| |
Collapse
|
7
|
Abstract
Mitochondria are involved in multiple cellular tasks, such as ATP synthesis, metabolism, metabolite and ion transport, regulation of apoptosis, inflammation, signaling, and inheritance of mitochondrial DNA. The majority of the correct functioning of mitochondria is based on the large electrochemical proton gradient, whose component, the inner mitochondrial membrane potential, is strictly controlled by ion transport through mitochondrial membranes. Consequently, mitochondrial function is critically dependent on ion homeostasis, the disturbance of which leads to abnormal cell functions. Therefore, the discovery of mitochondrial ion channels influencing ion permeability through the membrane has defined a new dimension of the function of ion channels in different cell types, mainly linked to the important tasks that mitochondrial ion channels perform in cell life and death. This review summarizes studies on animal mitochondrial ion channels with special focus on their biophysical properties, molecular identity, and regulation. Additionally, the potential of mitochondrial ion channels as therapeutic targets for several diseases is briefly discussed.
Collapse
Affiliation(s)
- Ildiko Szabo
- Department of Biology, University of Padova, Italy;
| | - Adam Szewczyk
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland;
| |
Collapse
|
8
|
Bernardi P, Carraro M, Lippe G. The mitochondrial permeability transition: Recent progress and open questions. FEBS J 2022; 289:7051-7074. [PMID: 34710270 PMCID: PMC9787756 DOI: 10.1111/febs.16254] [Citation(s) in RCA: 84] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Accepted: 10/27/2021] [Indexed: 01/13/2023]
Abstract
Major progress has been made in defining the basis of the mitochondrial permeability transition, a Ca2+ -dependent permeability increase of the inner membrane that has puzzled mitochondrial research for almost 70 years. Initially considered an artefact of limited biological interest by most, over the years the permeability transition has raised to the status of regulator of mitochondrial ion homeostasis and of druggable effector mechanism of cell death. The permeability transition is mediated by opening of channel(s) modulated by matrix cyclophilin D, the permeability transition pore(s) (PTP). The field has received new impulse (a) from the hypothesis that the PTP may originate from a Ca2+ -dependent conformational change of F-ATP synthase and (b) from the reevaluation of the long-standing hypothesis that it originates from the adenine nucleotide translocator (ANT). Here, we provide a synthetic account of the structure of ANT and F-ATP synthase to discuss potential and controversial mechanisms through which they may form high-conductance channels; and review some intriguing findings from the wealth of early studies of PTP modulation that still await an explanation. We hope that this review will stimulate new experiments addressing the many outstanding problems, and thus contribute to the eventual solution of the puzzle of the permeability transition.
Collapse
Affiliation(s)
- Paolo Bernardi
- Department of Biomedical Sciences and CNR Neuroscience InstituteUniversity of PadovaItaly
| | - Michela Carraro
- Department of Biomedical Sciences and CNR Neuroscience InstituteUniversity of PadovaItaly
| | | |
Collapse
|
9
|
Das K, Basak M, Mahata T, Kumar M, Kumar D, Biswas S, Chatterjee S, Moniruzzaman M, Saha NC, Mondal K, Kumar P, Das P, Stewart A, Maity B. RGS11-CaMKII complex mediated redox control attenuates chemotherapy-induced cardiac fibrosis. Redox Biol 2022; 57:102487. [PMID: 36228439 PMCID: PMC9557029 DOI: 10.1016/j.redox.2022.102487] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 09/20/2022] [Indexed: 12/06/2022] Open
Abstract
Dose limiting cardiotoxicity remains a major limiting factor in the clinical use of several cancer chemotherapeutics including anthracyclines and the antimetabolite 5-fluorouracil (5-FU). Prior work has demonstrated that chemotherapeutics increase expression of R7 family regulator of G protein signaling (RGS) protein-binding partner Gβ5, which drives myocyte cytotoxicity. However, though several R7 family members are expressed in heart, the exact role of each protein in chemotherapy driven heart damage remains unclear. Here, we demonstrate that RGS11, downregulated in the human heart following chemotherapy exposure, possesses potent anti-apoptotic actions, in direct opposition to the actions of fellow R7 family member RGS6. RGS11 forms a direct complex with the apoptotic kinase CaMKII and stress responsive transcription factor ATF3 and acts to counterbalance the ability of CaMKII and ATF3 to trigger oxidative stress, mitochondrial dysfunction, cell death, and release of the cardiokine neuregulin-1 (NRG1), which mediates pathological intercommunication between myocytes and endothelial cells. Doxorubicin triggers RGS11 depletion in the murine myocardium, and cardiac-specific OE of RGS11 decreases doxorubicin-induced fibrosis, myocyte hypertrophy, apoptosis, oxidative stress, and cell loss and aids in the maintenance of left ventricular function. Conversely, RGS11 knockdown in heart promotes cardiac fibrosis associated with CaMKII activation and ATF3/NRG1 induction. Indeed, inhibition of CaMKII largely prevents the fibrotic remodeling resulting from cardiac RGS11 depletion underscoring the functional importance of the RGS11-CaMKII interaction in the pathogenesis of cardiac fibrosis. These data describe an entirely new role for RGS11 in heart and identify RGS11 as a potential new target for amelioration of chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Kiran Das
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India; Academy of Scientific and Innovative Research (AcSIR), India
| | - Madhuri Basak
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Tarun Mahata
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Manish Kumar
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Dinesh Kumar
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India
| | - Sayan Biswas
- Forensic Medicine, College of Medicine and Sagore Dutta Hospital, B.T. Road, Kamarhati, Kolkata, West Bengal, 700058, India
| | | | | | | | - Kausik Mondal
- Zoology, University of Kalyani, Nadia, West Bengal, 741235, India
| | - Pranesh Kumar
- Pharmaceutical Sciences, Aryakul College of Pharmacy & Research, Natkur, Aryakul College Road, Lucknow, Uttar Pradesh, 226002, India
| | - Priyadip Das
- Chemistry, SRM Institute of Science and Technology, Kattankulathur, Tamilnadu, 603203, India
| | - Adele Stewart
- Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Jupiter, FL, 33458, USA
| | - Biswanath Maity
- Centre of Biomedical Research, SGPGIMS Campus, Raebareli Road, Lucknow, Uttar Pradesh, 226014, India; Academy of Scientific and Innovative Research (AcSIR), India.
| |
Collapse
|
10
|
Sun M, Wang R, Xia R, Xia Z, Wu Z, Wang T. Amelioration of myocardial ischemia/reperfusion injury in diabetes: A narrative review of the mechanisms and clinical applications of dexmedetomidine. Front Pharmacol 2022; 13:949754. [PMID: 36120296 PMCID: PMC9470922 DOI: 10.3389/fphar.2022.949754] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 08/08/2022] [Indexed: 11/13/2022] Open
Abstract
Mechanisms contributing to the pathogenesis of myocardial ischemia-reperfusion (I/R) injury are complex and multifactorial. Many strategies have been developed to ameliorate myocardial I/R injuries based on these mechanisms. However, the cardioprotective effects of these strategies appear to diminish in diabetic states. Diabetes weakens myocardial responses to therapies by disrupting intracellular signaling pathways which may be responsible for enhancing cellular resistance to damage. Intriguingly, it was found that Dexmedetomidine (DEX), a potent and selective α2-adrenergic agonist, appears to have the property to reverse diabetes-related inhibition of most intervention-mediated myocardial protection and exert a protective effect. Several mechanisms were revealed to be involved in DEX’s protection in diabetic rodent myocardial I/R models, including PI3K/Akt and associated GSK-3β pathway stimulation, endoplasmic reticulum stress (ERS) alleviation, and apoptosis inhibition. In addition, DEX could attenuate diabetic myocardial I/R injury by up-regulating autophagy, reducing ROS production, and inhibiting the inflammatory response through HMGB1 pathways. The regulation of autonomic nervous function also appeared to be involved in the protective mechanisms of DEX. In the present review, the evidence and underlying mechanisms of DEX in ameliorating myocardial I/R injury in diabetes are summarized, and the potential of DEX for the treatment/prevention of myocardial I/R injury in diabetic patients is discussed.
Collapse
Affiliation(s)
- Meng Sun
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rong Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Rui Xia
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhengyuan Xia
- State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong, China
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Zhilin Wu
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhilin Wu, ; Tingting Wang,
| | - Tingting Wang
- Department of Anesthesiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Institute of Anesthesia and Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- *Correspondence: Zhilin Wu, ; Tingting Wang,
| |
Collapse
|
11
|
Limbach MN, Antevska A, Oluwatoba DS, Gray ALH, Carroll XB, Hoffmann CM, Wang X, Voehler MW, Steren CA, Do TD. Atomic View of Aqueous Cyclosporine A: Unpacking a Decades-Old Mystery. J Am Chem Soc 2022; 144:12602-12607. [PMID: 35786958 DOI: 10.1021/jacs.2c01743] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
An atomic view of a main aqueous conformation of cyclosporine A (CycA), an important 11-amino-acid macrocyclic immunosuppressant, is reported. For decades, it has been a grand challenge to determine the conformation of free CycA in an aqueous-like solution given its poor water solubility. Using a combination of X-ray and single-crystal neutron diffraction, we unambiguously resolve a unique conformer (A1) with a novel cis-amide between residues 11 and 1 and two water ligands that stabilize hydrogen bond networks. NMR spectroscopy and titration experiments indicate that the novel conformer is as abundant as the closed conformer in 90/10 (v/v) methanol/water and is the main conformer at 10/90 methanol/water. Five other conformers were also detected in 90/10 methanol/water, one in slow exchange with A1, another one in slow exchange with the closed form and three minor ones, one of which contains two cis amides Abu2-Sar3 and MeBmt1-MeVal11. These conformers help better understand the wide spectrum of membrane permeability observed for CycA analogues and, to some extent, the binding of CycA to protein targets.
Collapse
Affiliation(s)
- Miranda N Limbach
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Aleksandra Antevska
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Damilola S Oluwatoba
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Amber L H Gray
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Xian B Carroll
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Christina M Hoffmann
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37830, United States
| | - Xiaoping Wang
- Neutron Scattering Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee 37830, United States
| | - Markus W Voehler
- Department of Chemistry, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Carlos A Steren
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| | - Thanh D Do
- Department of Chemistry, University of Tennessee, Knoxville, Tennessee 37996, United States
| |
Collapse
|
12
|
Taghavi S, Abdullah S, Toraih E, Packer J, Drury RH, Aras OA, Kosowski EM, Cotton-Betteridge A, Karim M, Bitonti N, Shaheen F, Duchesne J, Jackson-Weaver O. Dimethyl malonate slows succinate accumulation and preserves cardiac function in a swine model of hemorrhagic shock. J Trauma Acute Care Surg 2022; 93:13-20. [PMID: 35234713 PMCID: PMC9232889 DOI: 10.1097/ta.0000000000003593] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
BACKGROUND Succinate (SI) is a citric acid cycle metabolite that accumulates in tissues during hemorrhagic shock (HS) due to electron transport chain uncoupling. Dimethyl malonate (DMM) is a competitive inhibitor of SI dehydrogenase, which has been shown to reduce SI accumulation and protect against reperfusion injury. Whether DMM can be therapeutic after severe HS is unknown. We hypothesized that DMM would prevent SI buildup during resuscitation (RES) in a swine model of HS, leading to better physiological recovery after RES. METHODS The carotid arteries of Yorkshire pigs were cannulated with a 5-Fr catheter. After placement of a Swan-Ganz catheter and femoral arterial line, the carotid catheters were opened and the animals were exsanguinated to a mean arterial pressure (MAP) of 45 mm. After 30 minutes in the shock state, the animals were resuscitated to a MAP of 60 mm using lactated ringers. A MAP above 60 mm was maintained throughout RES. One group received 10 mg/kg of DMM (n = 6), while the control received sham injections (n = 6). The primary end-point was SI levels. Secondary end-points included cardiac function and lactate. RESULTS Succinate levels increased from baseline to the 20-minute RES point in control, while the DMM cohort remained unchanged. The DMM group required less intravenous fluid to maintain a MAP above 60 (450.0 vs. 229.0 mL; p = 0.01). The DMM group had higher pulmonary capillary wedge pressure at the 20-minute and 40-minute RES points. The DMM group had better recovery of cardiac output and index during RES, while the control had no improvement. While lactate levels were similar, DMM may lead to increased ionized calcium levels. DISCUSSION Dimethyl malonate slows SI accumulation during HS and helps preserve cardiac filling pressures and function during RES. In addition, DMM may protect against depletion of ionized calcium. Dimethyl malonate may have therapeutic potential during HS.
Collapse
Affiliation(s)
- Sharven Taghavi
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Sarah Abdullah
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Eman Toraih
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Jacob Packer
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Robert H. Drury
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Oguz A.Z. Aras
- Tulane University School of Medicine, New Orleans, Louisiana
| | | | | | - Mardeen Karim
- Tulane University School of Medicine, New Orleans, Louisiana
| | | | - Farhana Shaheen
- Tulane University School of Medicine, New Orleans, Louisiana
| | - Juan Duchesne
- Tulane University School of Medicine, New Orleans, Louisiana
| | | |
Collapse
|
13
|
Lui A, Kumar KK, Grant GA. Management of Severe Traumatic Brain Injury in Pediatric Patients. FRONTIERS IN TOXICOLOGY 2022; 4:910972. [PMID: 35812167 PMCID: PMC9263560 DOI: 10.3389/ftox.2022.910972] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 05/10/2022] [Indexed: 11/23/2022] Open
Abstract
The optimal management of severe traumatic brain injury (TBI) in the pediatric population has not been well studied. There are a limited number of research articles studying the management of TBI in children. Given the prevalence of severe TBI in the pediatric population, it is crucial to develop a reference TBI management plan for this vulnerable population. In this review, we seek to delineate the differences between severe TBI management in adults and children. Additionally, we also discuss the known molecular pathogenesis of TBI. A better understanding of the pathophysiology of TBI will inform clinical management and development of therapeutics. Finally, we propose a clinical algorithm for the management and treatment of severe TBI in children using published data.
Collapse
Affiliation(s)
- Austin Lui
- Touro University College of Osteopathic Medicine, Vallejo, CA, United States
| | - Kevin K. Kumar
- Department of Neurosurgery, Stanford University, Stanford, CA, United States
- Division of Pediatric Neurosurgery, Lucile Packard Children’s Hospital, Palo Alto, CA, United States
| | - Gerald A. Grant
- Department of Neurosurgery, Stanford University, Stanford, CA, United States
- Division of Pediatric Neurosurgery, Lucile Packard Children’s Hospital, Palo Alto, CA, United States
- Department of Neurosurgery, Duke University, Durham, NC, United States
| |
Collapse
|
14
|
Vlasov AV, Osipov SD, Bondarev NA, Uversky VN, Borshchevskiy VI, Yanyushin MF, Manukhov IV, Rogachev AV, Vlasova AD, Ilyinsky NS, Kuklin AI, Dencher NA, Gordeliy VI. ATP synthase F OF 1 structure, function, and structure-based drug design. Cell Mol Life Sci 2022; 79:179. [PMID: 35253091 PMCID: PMC11072866 DOI: 10.1007/s00018-022-04153-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/26/2021] [Accepted: 01/14/2022] [Indexed: 11/30/2022]
Abstract
ATP synthases are unique rotatory molecular machines that supply biochemical reactions with adenosine triphosphate (ATP)-the universal "currency", which cells use for synthesis of vital molecules and sustaining life. ATP synthases of F-type (FOF1) are found embedded in bacterial cellular membrane, in thylakoid membranes of chloroplasts, and in mitochondrial inner membranes in eukaryotes. The main functions of ATP synthases are control of the ATP synthesis and transmembrane potential. Although the key subunits of the enzyme remain highly conserved, subunit composition and structural organization of ATP synthases and their assemblies are significantly different. In addition, there are hypotheses that the enzyme might be involved in the formation of the mitochondrial permeability transition pore and play a role in regulation of the cell death processes. Dysfunctions of this enzyme lead to numerous severe disorders with high fatality levels. In our review, we focus on FOF1-structure-based approach towards development of new therapies by using FOF1 structural features inherited by the representatives of this enzyme family from different taxonomy groups. We analyzed and systematized the most relevant information about the structural organization of FOF1 to discuss how this approach might help in the development of new therapies targeting ATP synthases and design tools for cellular bioenergetics control.
Collapse
Affiliation(s)
- Alexey V Vlasov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Joint Institute for Nuclear Research, 141980, Dubna, Russia
| | - Stepan D Osipov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
| | - Nikolay A Bondarev
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
| | - Vladimir N Uversky
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Department of Molecular Medicine and Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
| | - Valentin I Borshchevskiy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52425, Jülich, Germany
- JuStruct: Jülich Center for Structural Biology, Forschungszentrum Jülich, 52428, Jülich, Germany
| | - Mikhail F Yanyushin
- Institute of Basic Biological Problems, Russian Academy of Sciences, 142290, Pushchino, Moscow region, Russia
| | - Ilya V Manukhov
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
| | - Andrey V Rogachev
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Joint Institute for Nuclear Research, 141980, Dubna, Russia
| | - Anastasiia D Vlasova
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
| | - Nikolay S Ilyinsky
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
| | - Alexandr I Kuklin
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Joint Institute for Nuclear Research, 141980, Dubna, Russia
| | - Norbert A Dencher
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia
- Physical Biochemistry, Department Chemistry, Technische Universität Darmstadt, Alarich-Weiss-Straße 4, 64287, Darmstadt, Germany
| | - Valentin I Gordeliy
- Research Center for Molecular Mechanisms of Aging and Age-Related Diseases, Moscow Institute of Physics and Technology, 141700, Dolgoprudny, Russia.
- Institute of Biological Information Processing (IBI-7: Structural Biochemistry), Forschungszentrum Jülich, 52425, Jülich, Germany.
- JuStruct: Jülich Center for Structural Biology, Forschungszentrum Jülich, 52428, Jülich, Germany.
- Institut de Biologie Structurale Jean-Pierre Ebel, Université Grenoble Alpes-Commissariat à l'Energie Atomique et aux Energies Alternatives-CNRS, 38027, Grenoble, France.
| |
Collapse
|
15
|
Boyenle ID, Oyedele AK, Ogunlana AT, Adeyemo AF, Oyelere FS, Akinola OB, Adelusi TI, Ehigie LO, Ehigie AF. Targeting the mitochondrial permeability transition pore for drug discovery: Challenges and opportunities. Mitochondrion 2022; 63:57-71. [PMID: 35077882 DOI: 10.1016/j.mito.2022.01.006] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/22/2021] [Accepted: 01/17/2022] [Indexed: 12/29/2022]
Abstract
Several drug targets have been amenable to drug discovery pursuit not until the characterization of the mitochondrial permeability transition pore (MPTP), a pore with an undefined molecular identity that forms on the inner mitochondrial membrane upon mitochondrial permeability transition (MPT) under the influence of calcium overload and oxidative stress. The opening of the pore which is presumed to cause cell death in certain human diseases also has implications under physiological parlance. Different models for this pore have been postulated following its first identification in the last six decades. The mitochondrial community has witnessed many protein candidates such as; voltage-dependent anion channel (VDAC), adenine nucleotide translocase (ANT), Mitochondrial phosphate carrier (PiC), Spastic Paralegin (SPG7), disordered proteins, and F1Fo ATPase. However, genetic studies have cast out most of these candidates with only F1Fo ATPase currently under intense argument. Cyclophilin D (CyPD) remains the widely accepted positive regulator of the MPTP known to date, but no drug candidate has emerged as its inhibitor, raising concern issues for therapeutics. Thus, in this review, we discuss various models of MPTP reported with the hope of stimulating further research in this field. We went beyond the classical description of the MPTP to ascribe a 'two-edged sword property' to the pore for therapeutic function in human disease because its inhibition and activation have pharmacological relevance. We suggested putative proteins upstream to CyPD that can regulate its activity and prevent cell deaths in neurodegenerative disease and ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ibrahim Damilare Boyenle
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria; Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Abdulquddus Kehinde Oyedele
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Abdeen Tunde Ogunlana
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Aishat Folashade Adeyemo
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | | | - Olateju Balikis Akinola
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Temitope Isaac Adelusi
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Leonard Ona Ehigie
- Computational Biology/Drug Discovery Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria
| | - Adeola Folasade Ehigie
- Membrane Biochemistry and Biophysics Research Laboratory, Department of Biochemistry, Ladoke Akintola University of Technology, Ogbomoso, Nigeria.
| |
Collapse
|
16
|
Carrer A, Tommasin L, Šileikytė J, Ciscato F, Filadi R, Urbani A, Forte M, Rasola A, Szabò I, Carraro M, Bernardi P. Defining the molecular mechanisms of the mitochondrial permeability transition through genetic manipulation of F-ATP synthase. Nat Commun 2021; 12:4835. [PMID: 34376679 PMCID: PMC8355262 DOI: 10.1038/s41467-021-25161-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 07/26/2021] [Indexed: 12/13/2022] Open
Abstract
F-ATP synthase is a leading candidate as the mitochondrial permeability transition pore (PTP) but the mechanism(s) leading to channel formation remain undefined. Here, to shed light on the structural requirements for PTP formation, we test cells ablated for g, OSCP and b subunits, and ρ0 cells lacking subunits a and A6L. Δg cells (that also lack subunit e) do not show PTP channel opening in intact cells or patch-clamped mitoplasts unless atractylate is added. Δb and ΔOSCP cells display currents insensitive to cyclosporin A but inhibited by bongkrekate, suggesting that the adenine nucleotide translocator (ANT) can contribute to channel formation in the absence of an assembled F-ATP synthase. Mitoplasts from ρ0 mitochondria display PTP currents indistinguishable from their wild-type counterparts. In this work, we show that peripheral stalk subunits are essential to turn the F-ATP synthase into the PTP and that the ANT provides mitochondria with a distinct permeability pathway.
Collapse
Affiliation(s)
- Andrea Carrer
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ludovica Tommasin
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Justina Šileikytė
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Francesco Ciscato
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Riccardo Filadi
- Department of Biomedical Sciences, University of Padova, Padova, Italy.,Consiglio Nazionale delle Ricerche Neuroscience Institute, Padova, Italy
| | - Andrea Urbani
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Michael Forte
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Andrea Rasola
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ildikò Szabò
- Consiglio Nazionale delle Ricerche Neuroscience Institute, Padova, Italy.,Department of Biology, University of Padova, Padova, Italy
| | - Michela Carraro
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
| | - Paolo Bernardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy. .,Consiglio Nazionale delle Ricerche Neuroscience Institute, Padova, Italy.
| |
Collapse
|
17
|
Morciano G, Naumova N, Koprowski P, Valente S, Sardão VA, Potes Y, Rimessi A, Wieckowski MR, Oliveira PJ. The mitochondrial permeability transition pore: an evolving concept critical for cell life and death. Biol Rev Camb Philos Soc 2021; 96:2489-2521. [PMID: 34155777 DOI: 10.1111/brv.12764] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 05/28/2021] [Accepted: 06/04/2021] [Indexed: 02/06/2023]
Abstract
In this review, we summarize current knowledge of perhaps one of the most intriguing phenomena in cell biology: the mitochondrial permeability transition pore (mPTP). This phenomenon, which was initially observed as a sudden loss of inner mitochondrial membrane impermeability caused by excessive calcium, has been studied for almost 50 years, and still no definitive answer has been provided regarding its mechanisms. From its initial consideration as an in vitro artifact to the current notion that the mPTP is a phenomenon with physiological and pathological implications, a long road has been travelled. We here summarize the role of mitochondria in cytosolic calcium control and the evolving concepts regarding the mitochondrial permeability transition (mPT) and the mPTP. We show how the evolving mPTP models and mechanisms, which involve many proposed mitochondrial protein components, have arisen from methodological advances and more complex biological models. We describe how scientific progress and methodological advances have allowed milestone discoveries on mPTP regulation and composition and its recognition as a valid target for drug development and a critical component of mitochondrial biology.
Collapse
Affiliation(s)
- Giampaolo Morciano
- Maria Cecilia Hospital, GVM Care & Research, Via Corriera 1, Cotignola, Ravenna, 48033, Italy.,Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Via Fossato di Mortara 70, Ferrara, 44121, Italy
| | - Natalia Naumova
- Department of Cardiac Thoracic and Vascular Sciences and Public Health, University of Padua Medical School, Via Giustiniani 2, Padova, 35128, Italy
| | - Piotr Koprowski
- Laboratory of Intracellular Ion Channels, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, Warsaw, 02-093, Poland
| | - Sara Valente
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, 3060-197, Portugal
| | - Vilma A Sardão
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, 3060-197, Portugal
| | - Yaiza Potes
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, Warsaw, 02-093, Poland
| | - Alessandro Rimessi
- Department of Medical Sciences, Section of Experimental Medicine, Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Via Fossato di Mortara 70, Ferrara, 44121, Italy
| | - Mariusz R Wieckowski
- Laboratory of Mitochondrial Biology and Metabolism, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, Warsaw, 02-093, Poland
| | - Paulo J Oliveira
- CNC - Center for Neuroscience and Cell Biology, CIBB - Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC Biotech, Biocant Park, Cantanhede, 3060-197, Portugal
| |
Collapse
|
18
|
Galber C, Minervini G, Cannino G, Boldrin F, Petronilli V, Tosatto S, Lippe G, Giorgio V. The f subunit of human ATP synthase is essential for normal mitochondrial morphology and permeability transition. Cell Rep 2021; 35:109111. [PMID: 33979610 DOI: 10.1016/j.celrep.2021.109111] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 03/11/2021] [Accepted: 04/20/2021] [Indexed: 01/03/2023] Open
Abstract
The f subunit is localized at the base of the ATP synthase peripheral stalk. Its function in the human enzyme is poorly characterized. Because full disruption of its ATP5J2 gene with the CRISPR-Cas9 strategy in the HAP1 human model has been shown to cause alterations in the amounts of other ATP synthase subunits, here we investigated the role of the f subunit in HeLa cells by regulating its levels through RNA interference. We confirm the role of the f subunit in ATP synthase dimer stability and observe that its downregulation per se does not alter the amounts of the other enzyme subunits or ATP synthase synthetic/hydrolytic activity. We show that downregulation of the f subunit causes abnormal crista organization and decreases permeability transition pore (PTP) size, whereas its re-expression in f subunit knockdown cells rescues mitochondrial morphology and PTP-dependent swelling.
Collapse
Affiliation(s)
- Chiara Galber
- Department of Biomedical Sciences, University of Padova, Padova 35121, Italy; Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padova 35121, Italy
| | - Giovanni Minervini
- Department of Biomedical Sciences, University of Padova, Padova 35121, Italy
| | - Giuseppe Cannino
- Department of Biomedical Sciences, University of Padova, Padova 35121, Italy
| | | | - Valeria Petronilli
- Department of Biomedical Sciences, University of Padova, Padova 35121, Italy; Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padova 35121, Italy
| | - Silvio Tosatto
- Department of Biomedical Sciences, University of Padova, Padova 35121, Italy
| | - Giovanna Lippe
- Department of Medicine, University of Udine, Udine 33100, Italy
| | - Valentina Giorgio
- Department of Biomedical Sciences, University of Padova, Padova 35121, Italy; Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padova 35121, Italy; Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna 40126, Italy.
| |
Collapse
|
19
|
Boyman L, Greiser M, Lederer WJ. Calcium influx through the mitochondrial calcium uniporter holocomplex, MCU cx. J Mol Cell Cardiol 2021; 151:145-154. [PMID: 33147447 PMCID: PMC7880866 DOI: 10.1016/j.yjmcc.2020.10.015] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2020] [Revised: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022]
Abstract
Ca2+ flux into the mitochondrial matrix through the MCU holocomplex (MCUcx) has recently been measured quantitatively and with milliseconds resolution for the first time under physiological conditions in both heart and skeletal muscle. Additionally, the dynamic levels of Ca2+ in the mitochondrial matrix ([Ca2+]m) of cardiomyocytes were measured as it was controlled by the balance between influx of Ca2+ into the mitochondrial matrix through MCUcx and efflux through the mitochondrial Na+ / Ca2+ exchanger (NCLX). Under these conditions [Ca2+]m was shown to regulate ATP production by the mitochondria at only a few critical sites. Additional functions attributed to [Ca2+]m continue to be reported in the literature. Here we review the new findings attributed to MCUcx function and provide a framework for understanding and investigating mitochondrial Ca2+ influx features, many of which remain controversial. The properties and functions of the MCUcx subunits that constitute the holocomplex are challenging to tease apart. Such distinct subunits include EMRE, MCUR1, MICUx (i.e. MICU1, MICU2, MICU3), and the pore-forming subunits (MCUpore). Currently, the specific set of functions of each subunit remains non-quantitative and controversial. The more contentious issues are discussed in the context of the newly measured native MCUcx Ca2+ flux from heart and skeletal muscle. These MCUcx Ca2+ flux measurements have been shown to be a highly-regulated, tissue-specific with femto-Siemens Ca2+ conductances and with distinct extramitochondrial Ca2+ ([Ca2+]i) dependencies. These data from cardiac and skeletal muscle mitochondria have been examined quantitatively for their threshold [Ca2+]i levels and for hypothesized gatekeeping function and are discussed in the context of model cell (e.g. HeLa, MEF, HEK293, COS7 cells) measurements. Our new findings on MCUcx dependent matrix [Ca2+]m signaling provide a quantitative basis for on-going and new investigations of the roles of MCUcx in cardiac function ranging from metabolic fuel selection, capillary blood-flow control and the pathological activation of the mitochondrial permeability transition pore (mPTP). Additionally, this review presents the use of advanced new methods that can be readily adapted by any investigator to enable them to carry out quantitative Ca2+ measurements in mitochondria while controlling the inner mitochondrial membrane potential, ΔΨm.
Collapse
Affiliation(s)
- Liron Boyman
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA; The Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Maura Greiser
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA; The Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - W Jonathan Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD, USA; The Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
20
|
Brustovetsky N. The Role of Adenine Nucleotide Translocase in the Mitochondrial Permeability Transition. Cells 2020; 9:E2686. [PMID: 33333766 PMCID: PMC7765165 DOI: 10.3390/cells9122686] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/10/2020] [Accepted: 12/12/2020] [Indexed: 12/30/2022] Open
Abstract
The mitochondrial permeability transition, a Ca2+-induced significant increase in permeability of the inner mitochondrial membrane, plays an important role in various pathologies. The mitochondrial permeability transition is caused by induction of the permeability transition pore (PTP). Despite significant effort, the molecular composition of the PTP is not completely clear and remains an area of hot debate. The Ca2+-modified adenine nucleotide translocase (ANT) and F0F1 ATP synthase are the major contenders for the role of pore in the PTP. This paper briefly overviews experimental results focusing on the role of ANT in the mitochondrial permeability transition and proposes that multiple molecular entities might be responsible for the conductance pathway of the PTP. Consequently, the term PTP cannot be applied to a single specific protein such as ANT or a protein complex such as F0F1 ATP synthase, but rather should comprise a variety of potential contributors to increased permeability of the inner mitochondrial membrane.
Collapse
Affiliation(s)
- Nickolay Brustovetsky
- Department of Pharmacology and Toxicology, Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
21
|
Amodeo GF, Pavlov EV. Amyloid β, α-synuclein and the c subunit of the ATP synthase: Can these peptides reveal an amyloidogenic pathway of the permeability transition pore? BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2020; 1863:183531. [PMID: 33309700 DOI: 10.1016/j.bbamem.2020.183531] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/26/2020] [Accepted: 11/09/2020] [Indexed: 01/29/2023]
Abstract
Mitochondrial Permeability Transition (PT) is a phenomenon of increased permeability of the inner mitochondrial membrane in response to high levels of Ca2+ and/or reactive oxygen species (ROS) in the matrix. PT occurs upon the opening of a pore, namely the permeability transition pore (PTP), which dissipates the membrane potential uncoupling the respiratory chain. mPT activation and PTP formation can occur through multiple molecular pathways. The specific focus of this review is to discuss the possible molecular mechanisms of PTP that involve the participation of mitochondrially targeted amyloid peptides Aβ, α-synuclein and c subunit of the ATP synthase (ATPase). As activators of PTP, amyloid peptides are uniquely different from other activators because they are capable of forming channels in lipid bilayers. This property rises the possibility that in this permeabilization pathway the formation of the channel involves the direct participation of peptides, making it uniquely different from other PTP induction mechanisms. In this pathway, a critical step of PTP activation involves the import of amyloidogenic peptides from the cytosol into the matrix. In the matrix these peptides, which would fold into α-helical structure in native conditions, interact with cyclophilin D (CypD) and upon stimulation by elevated ROS and/or the Ca2+ spontaneously misfold into β-sheet ion conducting pores, causing PTP opening.
Collapse
Affiliation(s)
- Giuseppe F Amodeo
- Department of Molecular Pathobiology, New York University, United States of America.
| | - Evgeny V Pavlov
- Department of Molecular Pathobiology, New York University, United States of America.
| |
Collapse
|
22
|
Bhatia V, Sharma S. Role of mitochondrial dysfunction, oxidative stress and autophagy in progression of Alzheimer's disease. J Neurol Sci 2020; 421:117253. [PMID: 33476985 DOI: 10.1016/j.jns.2020.117253] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2017] [Revised: 10/21/2020] [Accepted: 11/24/2020] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is the most common form of dementia. The pathological hallmarks of AD are amyloid plaques [aggregates of amyloid beta (A)] and neurofibrillary tangles (aggregates of tau protein). Growing evidence suggests that tau accumulation is pathologically more relevant to the development of neurodegeneration and cognitive decline in AD patients than A plaques. Mitochondrial damage plays an important role in AD. Mitochondrial damage has been related to amyloid-beta or tau pathology or to the presence of specific presenilin-1 mutations. Elevate reactive oxygen species/reactive nitrogen species production and defective mitochondrial dynamic balance has been suggested to be the reason as well as the consequence of AD related pathology. Oxidative stress is a prominent early event in the pathogenesis of AD and is therefore believed to contribute to tau hyperphosphorylation. Several studies have shown that the autophagy pathway in neurons is important under physiological and pathological conditions. Therefore, this pathway plays a crucial role for the degradation of endogenous soluble tau. However, the relationship between mitochondrial dysfunctioning, oxidative stress, autophagy dysregulation, and neuronal cell death in AD remains unclear. Here, we review the latest progress in AD, with a special emphasis on mitochondrial dysfunctioning, oxidative stress, and autophagy. We also discuss the interlink mechanism of these three factors in AD.
Collapse
Affiliation(s)
- Vandana Bhatia
- School of Pharmaceutical and Healthcare, CT University, Ludhiana, Punjab, India
| | - Saurabh Sharma
- School of Pharmaceutical Sciences, CT University, Ludhiana, Punjab, India.
| |
Collapse
|
23
|
Neginskaya MA, Strubbe JO, Amodeo GF, West BA, Yakar S, Bazil JN, Pavlov EV. The very low number of calcium-induced permeability transition pores in the single mitochondrion. J Gen Physiol 2020; 152:e202012631. [PMID: 32810269 PMCID: PMC7537349 DOI: 10.1085/jgp.202012631] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/22/2020] [Accepted: 07/24/2020] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial permeability transition (PT) is a phenomenon of stress-induced increase in nonspecific permeability of the mitochondrial inner membrane that leads to disruption of oxidative phosphorylation and cell death. Quantitative measurement of the membrane permeability increase during PT is critically important for understanding the PT's impact on mitochondrial function. The elementary unit of PT is a PT pore (PTP), a single channel presumably formed by either ATP synthase or adenine nucleotide translocator (ANT). It is not known how many channels are open in a single mitochondrion during PT, which makes it difficult to quantitatively estimate the overall degree of membrane permeability. Here, we used wide-field microscopy to record mitochondrial swelling and quantitatively measure rates of single-mitochondrion volume increase during PT-induced high-amplitude swelling. PT was quantified by calculating the rates of water flux responsible for measured volume changes. The total water flux through the mitochondrial membrane of a single mitochondrion during PT was in the range of (2.5 ± 0.4) × 10-17 kg/s for swelling in 2 mM Ca2+ and (1.1 ± 0.2) × 10-17 kg/s for swelling in 200 µM Ca2+. Under these experimental conditions, a single PTP channel with ionic conductance of 1.5 nS could allow passage of water at the rate of 0.65 × 10-17 kg/s. Thus, we estimate the integral ionic conductance of the whole mitochondrion during PT to be 5.9 ± 0.9 nS for 2 mM concentration of Ca2+ and 2.6 ± 0.4 nS for 200 µM of Ca2+. The number of PTPs per mitochondrion ranged from one to nine. Due to the uncertainties in PTP structure and model parameters, PTP count results may be slightly underestimated. However, taking into account that each mitochondrion has ∼15,000 copies of ATP synthases and ANTs, our data imply that PTP activation is a rare event that occurs only in a small subpopulation of these proteins.
Collapse
Affiliation(s)
- Maria A. Neginskaya
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY
| | - Jasiel O. Strubbe
- Department of Pharmacology and Toxicology, Michigan State University, East Lansing, MI
- Department of Physiology, Michigan State University, East Lansing, MI
| | - Giuseppe F. Amodeo
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY
| | - Benjamin A. West
- Department of Physiology, Michigan State University, East Lansing, MI
| | - Shoshana Yakar
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY
| | - Jason N. Bazil
- Department of Physiology, Michigan State University, East Lansing, MI
| | - Evgeny V. Pavlov
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY
| |
Collapse
|
24
|
Licznerski P, Park HA, Rolyan H, Chen R, Mnatsakanyan N, Miranda P, Graham M, Wu J, Cruz-Reyes N, Mehta N, Sohail S, Salcedo J, Song E, Effman C, Effman S, Brandao L, Xu GN, Braker A, Gribkoff VK, Levy RJ, Jonas EA. ATP Synthase c-Subunit Leak Causes Aberrant Cellular Metabolism in Fragile X Syndrome. Cell 2020; 182:1170-1185.e9. [PMID: 32795412 DOI: 10.1016/j.cell.2020.07.008] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2018] [Revised: 03/04/2020] [Accepted: 07/10/2020] [Indexed: 12/26/2022]
Abstract
Loss of the gene (Fmr1) encoding Fragile X mental retardation protein (FMRP) causes increased mRNA translation and aberrant synaptic development. We find neurons of the Fmr1-/y mouse have a mitochondrial inner membrane leak contributing to a "leak metabolism." In human Fragile X syndrome (FXS) fibroblasts and in Fmr1-/y mouse neurons, closure of the ATP synthase leak channel by mild depletion of its c-subunit or pharmacological inhibition normalizes stimulus-induced and constitutive mRNA translation rate, decreases lactate and key glycolytic and tricarboxylic acid (TCA) cycle enzyme levels, and triggers synapse maturation. FMRP regulates leak closure in wild-type (WT), but not FX synapses, by stimulus-dependent ATP synthase β subunit translation; this increases the ratio of ATP synthase enzyme to its c-subunit, enhancing ATP production efficiency and synaptic growth. In contrast, in FXS, inability to close developmental c-subunit leak prevents stimulus-dependent synaptic maturation. Therefore, ATP synthase c-subunit leak closure encourages development and attenuates autistic behaviors.
Collapse
Affiliation(s)
- Pawel Licznerski
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA.
| | - Han-A Park
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA; Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - Harshvardhan Rolyan
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Rongmin Chen
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Nelli Mnatsakanyan
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Paige Miranda
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Morven Graham
- Department of Cell Biology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jing Wu
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | | | - Nikita Mehta
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Sana Sohail
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Jorge Salcedo
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Erin Song
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | | | - Samuel Effman
- Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Lucas Brandao
- Department of Biology, Clark University, Worcester, MA 01610, USA
| | - Gulan N Xu
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Amber Braker
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA
| | - Valentin K Gribkoff
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA; Marine Biological Laboratory, Woods Hole, MA 02543, USA
| | - Richard J Levy
- Department of Anesthesiology, Columbia University Medical Center, New York, NY 10032, USA
| | - Elizabeth A Jonas
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT 06511, USA; Marine Biological Laboratory, Woods Hole, MA 02543, USA.
| |
Collapse
|
25
|
Mitochondrial F-ATP synthase as the permeability transition pore. Pharmacol Res 2020; 160:105081. [PMID: 32679179 DOI: 10.1016/j.phrs.2020.105081] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 07/06/2020] [Accepted: 07/10/2020] [Indexed: 12/27/2022]
Abstract
The current state of research on the mitochondrial permeability transition pore (PTP) can be described in terms of three major problems: molecular identity, atomic structure and gating mechanism. In this review these three problems are discussed in the light of recent findings with special emphasis on the discovery that the PTP is mitochondrial F-ATP synthase (mtFoF1). Novel features of the mitochondrial F-ATP synthase emerging from the success of single particle cryo electron microscopy (cryo-EM) to determine F-ATP synthase structures are surveyed along with their possible involvement in pore formation. Also, current findings from the gap junction field concerning the involvement of lipids in channel closure are examined. Finally, an earlier proposal denoted as the 'Death Finger' is discussed as a working model for PTP gating.
Collapse
|
26
|
Carraro M, Carrer A, Urbani A, Bernardi P. Molecular nature and regulation of the mitochondrial permeability transition pore(s), drug target(s) in cardioprotection. J Mol Cell Cardiol 2020; 144:76-86. [DOI: 10.1016/j.yjmcc.2020.05.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 04/28/2020] [Accepted: 05/20/2020] [Indexed: 12/12/2022]
|
27
|
Lamade AM, Anthonymuthu TS, Hier ZE, Gao Y, Kagan VE, Bayır H. Mitochondrial damage & lipid signaling in traumatic brain injury. Exp Neurol 2020; 329:113307. [PMID: 32289317 DOI: 10.1016/j.expneurol.2020.113307] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 04/09/2020] [Accepted: 04/10/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria are essential for neuronal function because they serve not only to sustain energy and redox homeostasis but also are harbingers of death. A dysregulated mitochondrial network can cascade until function is irreparably lost, dooming cells. TBI is most prevalent in the young and comes at significant personal and societal costs. Traumatic brain injury (TBI) triggers a biphasic and mechanistically heterogenous response and this mechanistic heterogeneity has made the development of standardized treatments challenging. The secondary phase of TBI injury evolves over hours and days after the initial insult, providing a window of opportunity for intervention. However, no FDA approved treatment for neuroprotection after TBI currently exists. With recent advances in detection techniques, there has been increasing recognition of the significance and roles of mitochondrial redox lipid signaling in both acute and chronic central nervous system (CNS) pathologies. Oxidized lipids and their downstream products result from and contribute to TBI pathogenesis. Therapies targeting the mitochondrial lipid composition and redox state show promise in experimental TBI and warrant further exploration. In this review, we provide 1) an overview for mitochondrial redox homeostasis with emphasis on glutathione metabolism, 2) the key mechanisms of TBI mitochondrial injury, 3) the pathways of mitochondria specific phospholipid cardiolipin oxidation, and 4) review the mechanisms of mitochondria quality control in TBI with consideration of the roles lipids play in this process.
Collapse
Affiliation(s)
- Andrew M Lamade
- Department of Critical Care Medicine, Safar Center for Resuscitation Research UPMC, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Tamil S Anthonymuthu
- Department of Critical Care Medicine, Safar Center for Resuscitation Research UPMC, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Zachary E Hier
- Department of Critical Care Medicine, Safar Center for Resuscitation Research UPMC, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Yuan Gao
- Department of Critical Care Medicine, Safar Center for Resuscitation Research UPMC, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA
| | - Valerian E Kagan
- Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA; Institute for Regenerative Medicine, IM Sechenov First Moscow State Medical University, Russian Federation
| | - Hülya Bayır
- Department of Critical Care Medicine, Safar Center for Resuscitation Research UPMC, Pittsburgh, PA, USA; Department of Environmental and Occupational Health, Center for Free Radical and Antioxidant Health, University of Pittsburgh, Pittsburgh, PA, USA; Children's Neuroscience Institute, UPMC Children's Hospital of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
28
|
ATP Synthase C-Subunit-Deficient Mitochondria Have a Small Cyclosporine A-Sensitive Channel, but Lack the Permeability Transition Pore. Cell Rep 2020; 26:11-17.e2. [PMID: 30605668 PMCID: PMC6521848 DOI: 10.1016/j.celrep.2018.12.033] [Citation(s) in RCA: 129] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 08/01/2018] [Accepted: 12/06/2018] [Indexed: 01/14/2023] Open
Abstract
Permeability transition (PT) is an increase in mitochondrial inner membrane permeability that can lead to a disruption of mitochondrial function and cell death. PT is responsible for tissue damage in stroke and myocardial infarction. It is caused by the opening of a large conductance (~1.5 nS) channel, the mitochondrial PT pore (mPTP). We directly tested the role of the c-subunit of ATP synthase in mPTP formation by measuring channel activity in c-subunit knockout mitochondria. We found that the classic mPTP conductance was lacking in c-subunit knockout mitochondria, but channels sensitive to the PT inhibitor cyclosporine A could be recorded. These channels had a significantly lower conductance compared with the cyclosporine A-sensitive channels detected in parental cells and were sensitive to the ATP/ADP translocase inhibitor bongkrekic acid. We propose that, in the absence of the c-subunit, mPTP cannot be formed, and a distinct cyclosporine A-sensitive low-conductance channel emerges. Neginskaya et al. report that c-subunit-deficient mitochondria contain a CSA-sensitive channel. This channel is much smaller compared with the wild-type permeability transition pore and is sensitive to inhibitors of adenine nucleotide translocase. This work highlights the importance of the c-subunit in forming the permeability transition pore.
Collapse
|
29
|
Wacquier B, Combettes L, Dupont G. Dual dynamics of mitochondrial permeability transition pore opening. Sci Rep 2020; 10:3924. [PMID: 32127570 PMCID: PMC7054270 DOI: 10.1038/s41598-020-60177-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2019] [Accepted: 02/06/2020] [Indexed: 11/09/2022] Open
Abstract
Mitochondria play an essential role in bioenergetics and cellular Ca\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${}^{2+}$$\end{document}2+ handling. The mitochondrial permeability transition pore (mPTP) is a non-specific channel located in the inner mitochondrial membrane. Long-lasting openings of the pore allow the rapid passage of ions and large molecules, which can result in cell death. The mPTP also exhibits transient, low conductance openings that contribute to Ca\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${}^{2+}$$\end{document}2+ homeostasis. Although many regulators of the pore have been identified, none of them uniquely governs the passage between the two operating modes, which thus probably relies on a still unidentified network of interactions. By developing a core computational model for mPTP opening under the control of mitochondrial voltage and Ca\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${}^{2+}$$\end{document}2+, we uncovered the existence of a positive feedback loop leading to bistability. The characteristics of the two stable steady-states correspond to those of the two opening states. When inserted in a full model of Ca\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${}^{2+}$$\end{document}2+ handling by mitochondria, our description of the pore reproduces observations in mitochondrial suspensions. Moreover, the model predicted the occurrence of hysteresis in the switching between the two modes, upon addition and removal of free Ca\documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}$${}^{2+}$$\end{document}2+ in the extra-mitochondrial medium. Stochastic simulations then confirmed that the pore can undergo transient openings resembling those observed in intact cells.
Collapse
Affiliation(s)
- Benjamin Wacquier
- Unit of Theoretical Chronobiology, Faculté des Sciences, Université Libre de Bruxelles (ULB) CP231, B1050, Brussels, Belgium
| | | | - Geneviève Dupont
- Unit of Theoretical Chronobiology, Faculté des Sciences, Université Libre de Bruxelles (ULB) CP231, B1050, Brussels, Belgium.
| |
Collapse
|
30
|
The Role of Mitochondria in the Mechanisms of Cardiac Ischemia-Reperfusion Injury. Antioxidants (Basel) 2019; 8:antiox8100454. [PMID: 31590423 PMCID: PMC6826663 DOI: 10.3390/antiox8100454] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 09/30/2019] [Accepted: 10/01/2019] [Indexed: 01/11/2023] Open
Abstract
Mitochondria play a critical role in maintaining cellular function by ATP production. They are also a source of reactive oxygen species (ROS) and proapoptotic factors. The role of mitochondria has been established in many aspects of cell physiology/pathophysiology, including cell signaling. Mitochondria may deteriorate under various pathological conditions, including ischemia-reperfusion (IR) injury. Mitochondrial injury can be one of the main causes for cardiac and other tissue injuries by energy stress and overproduction of toxic reactive oxygen species, leading to oxidative stress, elevated calcium and apoptotic and necrotic cell death. However, the interplay among these processes in normal and pathological conditions is still poorly understood. Mitochondria play a critical role in cardiac IR injury, where they are directly involved in several pathophysiological mechanisms. We also discuss the role of mitochondria in the context of mitochondrial dynamics, specializations and heterogeneity. Also, we wanted to stress the existence of morphologically and functionally different mitochondrial subpopulations in the heart that may have different sensitivities to diseases and IR injury. Therefore, various cardioprotective interventions that modulate mitochondrial stability, dynamics and turnover, including various pharmacologic agents, specific mitochondrial antioxidants and uncouplers, and ischemic preconditioning can be considered as the main strategies to protect mitochondrial and cardiovascular function and thus enhance longevity.
Collapse
|
31
|
Nesci S, Trombetti F, Algieri C, Pagliarani A. A Therapeutic Role for the F 1F O-ATP Synthase. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2019; 24:893-903. [PMID: 31266411 DOI: 10.1177/2472555219860448] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Recently, the F1FO-ATP synthase, due to its dual role of life enzyme as main adenosine triphosphate (ATP) maker and of death enzyme, as ATP dissipator and putative structural component of the mitochondrial permeability transition pore (mPTP), which triggers cell death, has been increasingly considered as a drug target. Accordingly, the enzyme offers new strategies to counteract the increased antibiotic resistance. The challenge is to find or synthesize compounds able to discriminate between prokaryotic and mitochondrial F1FO-ATP synthase, exploiting subtle structural differences to kill pathogens without affecting the host. From this perspective, the eukaryotic enzyme could also be made refractory to macrolide antibiotics by chemically produced posttranslational modifications. Moreover, because the mitochondrial F1FO-ATPase activity stimulated by Ca2+ instead of by the natural modulator Mg2+ is most likely involved in mPTP formation, effectors preferentially targeting the Ca2+-activated enzyme may modulate the mPTP. If the enzyme involvement in the mPTP is confirmed, Ca2+-ATPase inhibitors may counteract conditions featured by an increased mPTP activity, such as neurodegenerative and cardiovascular diseases and physiological aging. Conversely, mPTP opening could be pharmacologically stimulated to selectively kill unwanted cells. On the basis of recent literature and promising lab findings, the action mechanism of F1 and FO inhibitors is considered. These molecules may act as enzyme modifiers and constitute new drugs to kill pathogens, improve compromised enzyme functions, and limit the deathly enzyme role in pathologies. The enzyme offers a wide spectrum of therapeutic strategies to fight at the molecular level diseases whose treatment is still insufficient or merely symptomatic.
Collapse
Affiliation(s)
- Salvatore Nesci
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Bologna, Italy
| | - Fabiana Trombetti
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Bologna, Italy
| | - Cristina Algieri
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Bologna, Italy
| | - Alessandra Pagliarani
- Department of Veterinary Medical Sciences, University of Bologna, Ozzano Emilia, Bologna, Italy
| |
Collapse
|
32
|
Carraro M, Checchetto V, Szabó I, Bernardi P. F‐ATPsynthase and the permeability transition pore: fewer doubts, more certainties. FEBS Lett 2019; 593:1542-1553. [DOI: 10.1002/1873-3468.13485] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 06/09/2019] [Accepted: 06/10/2019] [Indexed: 12/27/2022]
Affiliation(s)
- Michela Carraro
- Department of Biomedical Sciences University of Padova Italy
| | | | - Ildikó Szabó
- Department of Biology University of Padova Italy
| | - Paolo Bernardi
- Department of Biomedical Sciences University of Padova Italy
| |
Collapse
|
33
|
Contribution of Mitochondrial Ion Channels to Chemo-Resistance in Cancer Cells. Cancers (Basel) 2019; 11:cancers11060761. [PMID: 31159324 PMCID: PMC6627730 DOI: 10.3390/cancers11060761] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 05/16/2019] [Accepted: 05/23/2019] [Indexed: 12/21/2022] Open
Abstract
Mitochondrial ion channels are emerging oncological targets, as modulation of these ion-transporting proteins may impact on mitochondrial membrane potential, efficiency of oxidative phosphorylation and reactive oxygen production. In turn, these factors affect the release of cytochrome c, which is the point of no return during mitochondrial apoptosis. Many of the currently used chemotherapeutics induce programmed cell death causing damage to DNA and subsequent activation of p53-dependent pathways that finally leads to cytochrome c release from the mitochondrial inter-membrane space. The view is emerging, as summarized in the present review, that ion channels located in this organelle may account in several cases for the resistance that cancer cells can develop against classical chemotherapeutics, by preventing drug-induced apoptosis. Thus, pharmacological modulation of these channel activities might be beneficial to fight chemo-resistance of different types of cancer cells.
Collapse
|
34
|
Shehadeh M, Palzur E, Apel L, Soustiel JF. Reduction of Traumatic Brain Damage by Tspo Ligand Etifoxine. Int J Mol Sci 2019; 20:ijms20112639. [PMID: 31146356 PMCID: PMC6600152 DOI: 10.3390/ijms20112639] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Revised: 05/25/2019] [Accepted: 05/26/2019] [Indexed: 01/30/2023] Open
Abstract
Experimental studies have shown that ligands of the 18 kDa translocator protein can reduce neuronal damage induced by traumatic brain injury by protecting mitochondria and preventing metabolic crisis. Etifoxine, an anxiolytic drug and 18 kDa translocator protein ligand, has shown beneficial effects in the models of peripheral nerve neuropathy. The present study investigates the potential effect of etifoxine as a neuroprotective agent in traumatic brain injury (TBI). For this purpose, the effect of etifoxine on lesion volume and modified neurological severity score at 4 weeks was tested in Sprague-Dawley adult male rats submitted to cortical impact contusion. Effects of etifoxine treatment on neuronal survival and apoptosis were also assessed by immune stains in the perilesional area. Etifoxine induced a significant reduction in the lesion volume compared to nontreated animals in a dose-dependent fashion with a similar effect on neurological outcome at four weeks that correlated with enhanced neuron survival and reduced apoptotic activity. These results are consistent with the neuroprotective effect of etifoxine in TBI that may justify further translational research.
Collapse
Affiliation(s)
- Mona Shehadeh
- Eliachar Research Laboratory, Galilee Medical Center, P.O. Box 21, Nahariya 2210001, Israel.
| | - Eilam Palzur
- Eliachar Research Laboratory, Galilee Medical Center, P.O. Box 21, Nahariya 2210001, Israel.
| | - Liat Apel
- Institute of Pathology, Galilee Medical Center, P.O. Box 21, Nahariya 2210001, Israel.
- The Azrieli Faculty of Medicine in the Galilee, Bar Ilan University, Safed 13100, Israel.
| | - Jean Francois Soustiel
- Eliachar Research Laboratory, Galilee Medical Center, P.O. Box 21, Nahariya 2210001, Israel.
- The Azrieli Faculty of Medicine in the Galilee, Bar Ilan University, Safed 13100, Israel.
- Department of Neurosurgery, Galilee Medical Center, P.O. Box 21, Nahariya 2210001, Israel.
| |
Collapse
|
35
|
Boyman L, Coleman AK, Zhao G, Wescott AP, Joca HC, Greiser BM, Karbowski M, Ward CW, Lederer WJ. Dynamics of the mitochondrial permeability transition pore: Transient and permanent opening events. Arch Biochem Biophys 2019; 666:31-39. [PMID: 30930285 PMCID: PMC6538282 DOI: 10.1016/j.abb.2019.03.016] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2018] [Revised: 03/19/2019] [Accepted: 03/26/2019] [Indexed: 12/11/2022]
Abstract
A gentle optical examination of the mitochondrial permeability transition pore (mPTP) opening events was carried out in isolated quiescent ventricular myocytes by tracking the inner membrane potential (ΔΨM) using TMRM (tetramethylrhodamine methyl ester). Zeiss Airyscan 880 ″super-resolution" or "high-resolution" imaging was done with very low levels of illumination (0.009% laser power). In cellular areas imaged every 9 s (ROI or regions of interest), transient depolarizations of variable amplitudes occurred at increasing rates for the first 30 min. The time to first depolarization events was 8.4 min (±1.1 SEM n = 21 cells). At longer times, essentially permanent and irreversible depolarizations occurred at an increasing fraction of all events. In other cellular areas surrounding the ROI, mitochondria were rarely illuminated (once per 5 min) and virtually no permanent depolarization events occurred for over 1 h of imaging. These findings suggest that photon stress due to the imaging itself plays an important role in the generation of both the transient mPTP opening events as well as the permanent mPTP opening events. Consistent with the evidence that photon "stress" in mitochondria loaded with virtually any photon absorbing substance, generates reactive oxygen species (ROS) [1-5], we show that cyclosporine-A (CsA, 10 μM) and the antioxidant n-acetyl cysteine (NAC, 10 mM), reduced the number of events by 80% and 93% respectively. Furthermore, CsA and NAC treatment led to the virtual disappearance of permanent depolarization events. Nevertheless, all transient depolarization events in any condition (control, CsA and NAC) appeared to repolarize with a similar half-time of 30 ± 6 s (n = 478) at 37 °C. Further experiments showed quantitatively similar results in cerebral vascular smooth muscle cells, using a different confocal system, and different photon absorbing reagent (TMRE; tetramethylrhodamine ethyl ester). In these experiments, using modest power (1% laser power) transient depolarization events were seen in only 8 out of 23 cells while with higher power (8%), all cells showed transient events, which align with the level of photon stress being the driver of the effect. Together, our findings suggest that photon-induced ROS is sufficient to cause depolarization events of individual mitochondria in quiescent cells; without electrical or mechanical activity to stimulates mitochondrial metabolism, and without raising the mitochondrial matrix Ca2+. In a broad context, these findings neither support nor deny the relevance or occurrence of ΔΨM depolarization events in specific putatively physiologic mitochondrial behaviors such as MitoFlashes [6,7] or MitoWinks [8]. Instead, our findings raise a caution with regards to the physiological and pathophysiological functions attributed to singular ΔΨM depolarization events when those functions are investigated using photon absorbing substances. Nevertheless, using photon stress as a tool ("Optical Stress-Probe"), we can extract information on the activation, reversibility, permanency and kinetics of mitochondrial depolarization. These data may provide new information on mPTP, help identify the mPTP protein complex, and establish the physiological function of the mPTP protein complex and their links to MitoFlashes and MitoWinks.
Collapse
Affiliation(s)
- Liron Boyman
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA; Department of Physiology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA.
| | - Andrew K Coleman
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA; Department of Physiology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA
| | - Guiling Zhao
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA; Department of Physiology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA
| | - Andrew P Wescott
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA; Department of Physiology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA
| | - Humberto C Joca
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA; Department of Physiology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA
| | - B Maura Greiser
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA; Department of Physiology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA
| | - Mariusz Karbowski
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA; Department of Biochemistry and Molecular Biology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA
| | - Chris W Ward
- Department of Orthopedics, University of Maryland School of Medicine, Baltimore, MD, USA
| | - W J Lederer
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA; Department of Physiology, University of Maryland School of Medicine, 111 Penn Street, Baltimore, MD, 21201, USA.
| |
Collapse
|
36
|
Leanza L, Checchetto V, Biasutto L, Rossa A, Costa R, Bachmann M, Zoratti M, Szabo I. Pharmacological modulation of mitochondrial ion channels. Br J Pharmacol 2019; 176:4258-4283. [PMID: 30440086 DOI: 10.1111/bph.14544] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/15/2018] [Accepted: 10/22/2018] [Indexed: 12/17/2022] Open
Abstract
The field of mitochondrial ion channels has undergone a rapid development during the last three decades, due to the molecular identification of some of the channels residing in the outer and inner membranes. Relevant information about the function of these channels in physiological and pathological settings was gained thanks to genetic models for a few, mitochondria-specific channels. However, many ion channels have multiple localizations within the cell, hampering a clear-cut determination of their function by pharmacological means. The present review summarizes our current knowledge about the ins and outs of mitochondrial ion channels, with special focus on the channels that have received much attention in recent years, namely, the voltage-dependent anion channels, the permeability transition pore (also called mitochondrial megachannel), the mitochondrial calcium uniporter and some of the inner membrane-located potassium channels. In addition, possible strategies to overcome the difficulties of specifically targeting mitochondrial channels versus their counterparts active in other membranes are discussed, as well as the possibilities of modulating channel function by small peptides that compete for binding with protein interacting partners. Altogether, these promising tools along with large-scale chemical screenings set up to identify new, specific channel modulators will hopefully allow us to pinpoint the actual function of most mitochondrial ion channels in the near future and to pharmacologically affect important pathologies in which they are involved, such as neurodegeneration, ischaemic damage and cancer. LINKED ARTICLES: This article is part of a themed section on Mitochondrial Pharmacology: Featured Mechanisms and Approaches for Therapy Translation. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v176.22/issuetoc.
Collapse
Affiliation(s)
- Luigi Leanza
- Department of Biology, University of Padova, Padova, Italy
| | | | - Lucia Biasutto
- CNR Institute of Neurosciences, Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Andrea Rossa
- Department of Chemical Sciences, University of Padova, Padova, Italy
| | - Roberto Costa
- Department of Biology, University of Padova, Padova, Italy
| | | | - Mario Zoratti
- CNR Institute of Neurosciences, Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Ildiko Szabo
- Department of Biology, University of Padova, Padova, Italy.,CNR Institute of Neurosciences, Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
37
|
Lemasters JJ, Zhong Z. Mitophagy in hepatocytes: Types, initiators and role in adaptive ethanol metabolism☆. LIVER RESEARCH 2018; 2:125-132. [PMID: 31157120 PMCID: PMC6541449 DOI: 10.1016/j.livres.2018.09.005] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Mitophagy (mitochondrial autophagy) in hepatocytes is an essential quality control mechanism that removes for lysosomal digestion damaged, effete and superfluous mitochondria. Mitophagy has distinct variants. In type 1 mitophagy, typical of nutrient deprivation, cup-shaped sequestration membranes (phagophores) grow, surround and sequester individual mitochondria into mitophagosomes, often in coordination with mitochondrial fission. After sequestration, the outer compartment of the mitophagosome acidifies and the entrapped mitochondrion depolarizes, followed by fusion with lysosomes. By contrast, mitochondrial depolarization stimulates type 2 mitophagy, which is characterized by coalescence of autophagic microtubule-associated protein 1A/1B-light chain 3 (LC3)-containing structures on mitochondrial surfaces without the formation of a phagophore or mitochondrial fission. Oppositely to type 1 mitophagy, the inhibition of phosphoinositide-3-kinase (PI3K) does not block type 2 mitophagy. In type 3 mitophagy, or micromitophagy, mitochondria-derived vesicles (MDVs) enriched in oxidized proteins bud off from mitochondrial inner and outer membranes and incorporate into multivesicular bodies by vesicle scission into the lumen. In response to ethanol feeding, widespread ethanol-induced hepatocellular mitochondrial depolarization occurs to facilitate hepatic ethanol metabolism. As a consequence, type 2 mitophagy develops in response to the mitochondrial depolarization. After chronic high ethanol feeding, processing of depolarized mitochondria by mitophagy becomes compromised, leading to release of mitochondrial damage-associated molecular patterns (mtDAMPs) that promote inflammatory and profibrogenic responses. We propose that the persistence of mitochondrial responses for acute ethanol metabolism links initial adaptive ethanol metabolism to mitophagy and then to chronic maladaptive changes initiating onset and the progression of alcoholic liver disease (ALD).
Collapse
Affiliation(s)
- John J. Lemasters
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
- Department of Biochemistry & Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | - Zhi Zhong
- Department of Drug Discovery & Biomedical Sciences, Medical University of South Carolina, Charleston, SC, USA
| |
Collapse
|
38
|
Calcineurin Silencing in Dictyostelium discoideum Leads to Cellular Alterations Affecting Mitochondria, Gene Expression, and Oxidative Stress Response. Protist 2018; 169:584-602. [DOI: 10.1016/j.protis.2018.04.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 04/04/2018] [Accepted: 04/11/2018] [Indexed: 11/18/2022]
|
39
|
Kamei Y, Koushi M, Aoyama Y, Asakai R. The yeast mitochondrial permeability transition is regulated by reactive oxygen species, endogenous Ca 2+ and Cpr3, mediating cell death. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:1313-1326. [PMID: 30031690 DOI: 10.1016/j.bbabio.2018.07.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 11/29/2022]
Abstract
We investigated the properties of the permeability transition pore (PTP) in Saccharomyces cerevisiae in agar-embedded mitochondria (AEM) and agar-embedded cells (AEC) and its role in yeast death. In AEM, ethanol-induced pore opening, as indicated by the release of calcein and mitochondrial membrane depolarization, can be inhibited by CsA, by Cpr3 deficiency, and by the antioxidant glutathione. Notably, the pore opening is inhibited, when mitochondria are preloaded by EGTA or Fluo3 to chelate matrix Ca2+, or are pretreated with 4-Br A23187 to extract matrix Ca2+, prior to agar-embedding, or when pore opening is induced in the presence of EGTA; opened pores are re-closed by sequential treatment with CsA, 4-Br A23187 plus EGTA and NADH, indicating endogenous matrix Ca2+ involvement. CsA also inhibits the pore opening with low conductance triggered by exogenous Ca2+ transport with ETH129. In AEC, the treatment of tert-butylhydroperoxide, a pro-oxidant that triggers transient pore opening in high conductance in AEM, induces yeast death, which is also dependent on CsA and Cpr3. Furthermore, AEMs from mutants lacking three ADP/ATP carrier (AAC) isoforms and with defective ATP synthase dimerization exhibit high and low conductance pore openings with CsA sensitivity, respectively. Collectively, these data show that the yeast PTP is regulated by Cpr3, endogenous matrix Ca2+, and reactive oxygen species, and that it is involved in yeast death; furthermore, ATP synthase dimers play a key role in CsA-sensitive pore formation, while AACs are dispensable.
Collapse
Affiliation(s)
- Yoshiko Kamei
- Department of Morphophysiology, Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba 283-8555, Japan
| | - Masami Koushi
- Department of Morphophysiology, Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba 283-8555, Japan
| | - Yasunori Aoyama
- Department of Morphophysiology, Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba 283-8555, Japan
| | - Rei Asakai
- Department of Morphophysiology, Faculty of Pharmaceutical Sciences, Josai International University, 1 Gumyo, Togane, Chiba 283-8555, Japan.
| |
Collapse
|
40
|
Bachmann M, Costa R, Peruzzo R, Prosdocimi E, Checchetto V, Leanza L. Targeting Mitochondrial Ion Channels to Fight Cancer. Int J Mol Sci 2018; 19:ijms19072060. [PMID: 30011966 PMCID: PMC6073807 DOI: 10.3390/ijms19072060] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 07/12/2018] [Accepted: 07/13/2018] [Indexed: 12/14/2022] Open
Abstract
In recent years, several experimental evidences have underlined a new role of ion channels in cancer development and progression. In particular, mitochondrial ion channels are arising as new oncological targets, since it has been proved that most of them show an altered expression during tumor development and the pharmacological targeting of some of them have been demonstrated to be able to modulate cancer growth and progression, both in vitro as well as in vivo in pre-clinical mouse models. In this scenario, pharmacology of mitochondrial ion channels would be in the near future a new frontier for the treatment of tumors. In this review, we discuss the new advances in the field, by focusing our attention on the improvements in new drug developments to target mitochondrial ion channels.
Collapse
Affiliation(s)
| | - Roberto Costa
- Department of Biology, University of Padova, 35131 Padova, Italy.
| | - Roberta Peruzzo
- Department of Biology, University of Padova, 35131 Padova, Italy.
| | - Elena Prosdocimi
- Department of Biology, University of Padova, 35131 Padova, Italy.
| | | | - Luigi Leanza
- Department of Biology, University of Padova, 35131 Padova, Italy.
| |
Collapse
|
41
|
Burstein SR, Kim HJ, Fels JA, Qian L, Zhang S, Zhou P, Starkov AA, Iadecola C, Manfredi G. Estrogen receptor beta modulates permeability transition in brain mitochondria. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2018; 1859:423-433. [PMID: 29550215 PMCID: PMC5912174 DOI: 10.1016/j.bbabio.2018.03.006] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 03/08/2018] [Accepted: 03/12/2018] [Indexed: 12/24/2022]
Abstract
Recent evidence highlights a role for sex and hormonal status in regulating cellular responses to ischemic brain injury and neurodegeneration. A key pathological event in ischemic brain injury is the opening of a mitochondrial permeability transition pore (MPT) induced by excitotoxic calcium levels, which can trigger irreversible damage to mitochondria accompanied by the release of pro-apoptotic factors. However, sex differences in brain MPT modulation have not yet been explored. Here, we show that mitochondria isolated from female mouse forebrain have a lower calcium threshold for MPT than male mitochondria, and that this sex difference depends on the MPT regulator cyclophilin D (CypD). We also demonstrate that an estrogen receptor beta (ERβ) antagonist inhibits MPT and knockout of ERβ decreases the sensitivity of mitochondria to the CypD inhibitor, cyclosporine A. These results suggest a functional relationship between ERβ and CypD in modulating brain MPT. Moreover, co-immunoprecipitation studies identify several ERβ binding partners in mitochondria. Among these, we investigate the mitochondrial ATPase as a putative site of MPT regulation by ERβ. We find that previously described interaction between the oligomycin sensitivity-conferring subunit of ATPase (OSCP) and CypD is decreased by ERβ knockout, suggesting that ERβ modulates MPT by regulating CypD interaction with OSCP. Functionally, in primary neurons and hippocampal slice cultures, modulation of ERβ has protective effects against glutamate toxicity and oxygen glucose deprivation, respectively. Taken together, these results reveal a novel pathway of brain MPT regulation by ERβ that could contribute to sex differences in ischemic brain injury and neurodegeneration.
Collapse
Affiliation(s)
- Suzanne R Burstein
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Hyun Jeong Kim
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Jasmine A Fels
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA; Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, 1300 York Avenue, New York, NY 10021, USA
| | - Liping Qian
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Sheng Zhang
- Proteomics and Mass Spectrometry Facility, 139 Biotechnology Building, Cornell University, 526 Campus Road, Ithaca, NY 14853, USA
| | - Ping Zhou
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Anatoly A Starkov
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA
| | - Giovanni Manfredi
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, 407 East 61st Street, New York, NY 10065, USA.
| |
Collapse
|
42
|
Antoniel M, Jones K, Antonucci S, Spolaore B, Fogolari F, Petronilli V, Giorgio V, Carraro M, Di Lisa F, Forte M, Szabó I, Lippe G, Bernardi P. The unique histidine in OSCP subunit of F-ATP synthase mediates inhibition of the permeability transition pore by acidic pH. EMBO Rep 2018; 19:257-268. [PMID: 29217657 PMCID: PMC5797955 DOI: 10.15252/embr.201744705] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2017] [Revised: 11/09/2017] [Accepted: 11/14/2017] [Indexed: 01/19/2023] Open
Abstract
The permeability transition pore (PTP) is a Ca2+-dependent mitochondrial channel whose opening causes a permeability increase in the inner membrane to ions and solutes. The most potent inhibitors are matrix protons, with channel block at pH 6.5. Inhibition is reversible, mediated by histidyl residue(s), and prevented by their carbethoxylation by diethylpyrocarbonate (DPC), but their assignment is unsolved. We show that PTP inhibition by H+ is mediated by the highly conserved histidyl residue (H112 in the human mature protein) of oligomycin sensitivity conferral protein (OSCP) subunit of mitochondrial F1FO (F)-ATP synthase, which we also show to undergo carbethoxylation after reaction of mitochondria with DPC. Mitochondrial PTP-dependent swelling cannot be inhibited by acidic pH in H112Q and H112Y OSCP mutants, and the corresponding megachannels (the electrophysiological counterpart of the PTP) are insensitive to inhibition by acidic pH in patch-clamp recordings of mitoplasts. Cells harboring the H112Q and H112Y mutations are sensitized to anoxic cell death at acidic pH. These results demonstrate that PTP channel formation and its inhibition by H+ are mediated by the F-ATP synthase.
Collapse
Affiliation(s)
- Manuela Antoniel
- Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Kristen Jones
- Vollum Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - Salvatore Antonucci
- Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Barbara Spolaore
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Padova, Italy
| | - Federico Fogolari
- Department of Mathematics, Computer Sciences and Physics, University of Udine, Udine, Italy
| | - Valeria Petronilli
- Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Valentina Giorgio
- Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Michela Carraro
- Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Fabio Di Lisa
- Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Michael Forte
- Vollum Institute, Oregon Health and Sciences University, Portland, OR, USA
| | - Ildikó Szabó
- Department of Biology, University of Padova, Padova, Italy
| | - Giovanna Lippe
- Department of Agricultural, Food, Environmental and Animal Sciences, University of Udine, Udine, Italy
| | - Paolo Bernardi
- Consiglio Nazionale delle Ricerche Institute of Neuroscience, Padova, Italy
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| |
Collapse
|
43
|
Abstract
Mitochondrial ATP generation by oxidative phosphorylation combines the stepwise oxidation by the electron transport chain (ETC) of the reducing equivalents NADH and FADH2 with the generation of ATP by the ATP synthase. Recent studies show that the ATP synthase is not only essential for the generation of ATP but may also contribute to the formation of the mitochondrial permeability transition pore (PTP). We present a model, in which the PTP is located within the c-subunit ring in the Fo subunit of the ATP synthase. Opening of the PTP was long associated with uncoupling of the ETC and the initiation of programmed cell death. More recently, it was shown that PTP opening may serve a physiologic role: it can transiently open to regulate mitochondrial signaling in mature cells, and it is open in the embryonic mouse heart. This review will discuss how the ATP synthase paradoxically lies at the center of both ATP generation and cell death.
Collapse
|
44
|
Zorov DB, Popkov VA, Zorova LD, Vorobjev IA, Pevzner IB, Silachev DN, Zorov SD, Jankauskas SS, Babenko VA, Plotnikov EY. Mitochondrial Aging: Is There a Mitochondrial Clock? J Gerontol A Biol Sci Med Sci 2017; 72:1171-1179. [PMID: 27927758 DOI: 10.1093/gerona/glw184] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2016] [Accepted: 08/23/2016] [Indexed: 01/16/2023] Open
Abstract
Fragmentation (fission) of mitochondria, occurring in response to oxidative challenge, leads to heterogeneity in the mitochondrial population. It is assumed that fission provides a way to segregate mitochondrial content between the "young" and "old" phenotype, with the formation of mitochondrial "garbage," which later will be disposed. Fidelity of this process is the basis of mitochondrial homeostasis, which is disrupted in pathological conditions and aging. The asymmetry of the mitochondrial fission is similar to that of their evolutionary ancestors, bacteria, which also undergo an aging process. It is assumed that mitochondrial markers of aging are recognized by the mitochondrial quality control system, preventing the accumulation of dysfunctional mitochondria, which normally are subjected to disposal. Possibly, oncocytoma, with its abnormal proliferation of mitochondria occupying the entire cytoplasm, represents the case when segregation of damaged mitochondria is impaired during mitochondrial division. It is plausible that mitochondria contain a "clock" which counts the degree of mitochondrial senescence as the extent of flagging (by ubiquitination) of damaged mitochondria. Mitochondrial aging captures the essence of the systemic aging which must be analyzed. We assume that the mitochondrial aging mechanism is similar to the mechanism of aging of the immune system which we discuss in detail.
Collapse
Affiliation(s)
| | | | | | - Ivan A Vorobjev
- Biological Faculty, Lomonosov Moscow State University, Russia
| | | | | | | | | | | | | |
Collapse
|
45
|
Giorgio V, Guo L, Bassot C, Petronilli V, Bernardi P. Calcium and regulation of the mitochondrial permeability transition. Cell Calcium 2017; 70:56-63. [PMID: 28522037 DOI: 10.1016/j.ceca.2017.05.004] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 05/05/2017] [Accepted: 05/05/2017] [Indexed: 12/11/2022]
Abstract
Recent years have seen renewed interest in the permeability transition pore, a high conductance channel responsible for permeabilization of the inner mitochondrial membrane, a process that leads to depolarization and Ca2+ release. Transient openings may be involved in physiological Ca2+ homeostasis while long-lasting openings may trigger and/or execute cell death. In this review we specifically focus (i) on the hypothesis that the PTP forms from the F-ATP synthase and (ii) on the mechanisms through which Ca2+ can reversibly switch this energy-conserving nanomachine into an energy-dissipating device.
Collapse
Affiliation(s)
- Valentina Giorgio
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Italy
| | - Lishu Guo
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Italy
| | - Claudio Bassot
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Italy
| | - Valeria Petronilli
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Italy
| | - Paolo Bernardi
- Department of Biomedical Sciences and CNR Neuroscience Institute, University of Padova, Italy.
| |
Collapse
|
46
|
Mnatsakanyan N, Beutner G, Porter GA, Alavian KN, Jonas EA. Physiological roles of the mitochondrial permeability transition pore. J Bioenerg Biomembr 2017; 49:13-25. [PMID: 26868013 PMCID: PMC4981558 DOI: 10.1007/s10863-016-9652-1] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 02/04/2016] [Indexed: 01/01/2023]
Abstract
Neurons experience high metabolic demand during such processes as synaptic vesicle recycling, membrane potential maintenance and Ca2+ exchange/extrusion. The energy needs of these events are met in large part by mitochondrial production of ATP through the process of oxidative phosphorylation. The job of ATP production by the mitochondria is performed by the F1FO ATP synthase, a multi-protein enzyme that contains a membrane-inserted portion, an extra-membranous enzymatic portion and an extensive regulatory complex. Although required for ATP production by mitochondria, recent findings have confirmed that the membrane-confined portion of the c-subunit of the ATP synthase also houses a large conductance uncoupling channel, the mitochondrial permeability transition pore (mPTP), the persistent opening of which produces osmotic dysregulation of the inner mitochondrial membrane, uncoupling of oxidative phosphorylation and cell death. Recent advances in understanding the molecular components of mPTP and its regulatory mechanisms have determined that decreased uncoupling occurs in states of enhanced mitochondrial efficiency; relative closure of mPTP therefore contributes to cellular functions as diverse as cardiac development and synaptic efficacy.
Collapse
Affiliation(s)
- Nelli Mnatsakanyan
- Department Internal Medicine, Section of Endocrinology, Yale University, New Haven, CT, USA
| | - Gisela Beutner
- Department of Pediatrics (Cardiology), University of Rochester Medical Center, Rochester, NY, USA
| | - George A Porter
- Department of Pediatrics (Cardiology), University of Rochester Medical Center, Rochester, NY, USA
| | - Kambiz N Alavian
- Division of Brain Sciences, Department of Medicine, Imperial College London, London, UK
| | - Elizabeth A Jonas
- Department Internal Medicine, Section of Endocrinology, Yale University, New Haven, CT, USA.
| |
Collapse
|
47
|
Elustondo PA, Nichols M, Negoda A, Thirumaran A, Zakharian E, Robertson GS, Pavlov EV. Mitochondrial permeability transition pore induction is linked to formation of the complex of ATPase C-subunit, polyhydroxybutyrate and inorganic polyphosphate. Cell Death Discov 2016; 2:16070. [PMID: 27924223 PMCID: PMC5137186 DOI: 10.1038/cddiscovery.2016.70] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Revised: 08/10/2016] [Accepted: 08/19/2016] [Indexed: 12/25/2022] Open
Abstract
Mitochondrial permeability transition pore (mPTP) opening allows free movement of ions and small molecules leading to mitochondrial membrane depolarization and ATP depletion that triggers cell death. A multi-protein complex of the mitochondrial ATP synthase has an essential role in mPTP. However, the molecular identity of the central 'pore' part of mPTP complex is not known. A highly purified fraction of mammalian mitochondria containing C-subunit of ATPase (C-subunit), calcium, inorganic polyphosphate (polyP) and polyhydroxybutyrate (PHB) forms ion channels with properties that resemble the native mPTP. We demonstrate here that amount of this channel-forming complex dramatically increases in intact mitochondria during mPTP activation. This increase is inhibited by both Cyclosporine A, an inhibitor of mPTP and Ruthenium Red, an inhibitor of the Mitochondrial Calcium Uniporter. Similar increases in the amount of complex formation occurs in areas of mouse brain damaged by ischemia-reperfusion injury. These findings suggest that calcium-induced mPTP is associated with de novo assembly of a channel comprising C-subunit, polyP and PHB.
Collapse
Affiliation(s)
- P A Elustondo
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University , Halifax, NS, B3H 4R2 Canada
| | - M Nichols
- Departments of Psychiatry and Pharmacology, Brain Repair Centre, Faculty of Medicine Dalhousie University , Halifax, NS, B3H 4R2f Canada
| | - A Negoda
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University , Halifax, NS, B3H 4R2 Canada
| | - A Thirumaran
- Departments of Psychiatry and Pharmacology, Brain Repair Centre, Faculty of Medicine Dalhousie University , Halifax, NS, B3H 4R2f Canada
| | - E Zakharian
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine , 1 Illini Drive, Peoria, IL 61605, USA
| | - G S Robertson
- Departments of Psychiatry and Pharmacology, Brain Repair Centre, Faculty of Medicine Dalhousie University , Halifax, NS, B3H 4R2f Canada
| | - E V Pavlov
- Department of Physiology and Biophysics, Faculty of Medicine, Dalhousie University, Halifax, NS, B3H 4R2 Canada; Department of Basic Sciences, New York University, College of Dentistry, 345 East 24th Street, New York, NY 10010, USA
| |
Collapse
|
48
|
Effect of Cyclosporin A on the Viability of Hippocampal Cells Cultured under Conditions of Modeling of Alzheimer’s Disease. NEUROPHYSIOLOGY+ 2016. [DOI: 10.1007/s11062-016-9595-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
49
|
Yan S, Du F, Wu L, Zhang Z, Zhong C, Yu Q, Wang Y, Lue LF, Walker DG, Douglas JT, Yan SS. F1F0 ATP Synthase-Cyclophilin D Interaction Contributes to Diabetes-Induced Synaptic Dysfunction and Cognitive Decline. Diabetes 2016; 65:3482-3494. [PMID: 27554467 PMCID: PMC5079631 DOI: 10.2337/db16-0556] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 08/09/2016] [Indexed: 02/03/2023]
Abstract
Mitochondrial abnormalities are well known to cause cognitive decline. However, the underlying molecular basis of mitochondria-associated neuronal and synaptic dysfunction in the diabetic brain remains unclear. Here, using a mitochondrial single-channel patch clamp and cyclophilin D (CypD)-deficient mice (Ppif -/-) with streptozotocin-induced diabetes, we observed an increase in the probability of Ca2+-induced mitochondrial permeability transition pore (mPTP) opening in brain mitochondria of diabetic mice, which was further confirmed by mitochondrial swelling and cytochrome c release induced by Ca2+ overload. Diabetes-induced elevation of CypD triggers enhancement of F1F0 ATP synthase-CypD interaction, which in turn leads to mPTP opening. Indeed, in patients with diabetes, brain cypD protein levels were increased. Notably, blockade of the F1F0 ATP synthase-CypD interaction by CypD ablation protected against diabetes-induced mPTP opening, ATP synthesis deficits, oxidative stress, and mitochondria dysfunction. Furthermore, the absence of CypD alleviated deficits in synaptic plasticity, learning, and memory in diabetic mice. Thus, blockade of ATP synthase interaction with CypD provides a promising new target for therapeutic intervention in diabetic encephalopathy.
Collapse
Affiliation(s)
- Shijun Yan
- Department of Pharmacology & Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Fang Du
- Department of Pharmacology & Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Long Wu
- Department of Pharmacology & Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Zhihua Zhang
- Department of Pharmacology & Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Changjia Zhong
- Department of Pharmacology & Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Qing Yu
- Department of Pharmacology & Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Yongfu Wang
- Department of Pharmacology & Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Lih-Fen Lue
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ
| | - Douglas G Walker
- Neurodegenerative Disease Research Center, Biodesign Institute, Arizona State University, Tempe, AZ
| | - Justin T Douglas
- Nuclear Magnetic Resonance Laboratory, Molecular Structures Group, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Shirley ShiDu Yan
- Department of Pharmacology & Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS
| |
Collapse
|
50
|
Zulian A, Schiavone M, Giorgio V, Bernardi P. Forty years later: Mitochondria as therapeutic targets in muscle diseases. Pharmacol Res 2016; 113:563-573. [PMID: 27697642 DOI: 10.1016/j.phrs.2016.09.043] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2016] [Accepted: 09/29/2016] [Indexed: 11/22/2022]
Abstract
The hypothesis that mitochondrial dysfunction can be a general mechanism for cell death in muscle diseases is 40 years old. The key elements of the proposed pathogenetic sequence (cytosolic Ca2+ overload followed by excess mitochondrial Ca2+ uptake, functional and then structural damage of mitochondria, energy shortage, worsened elevation of cytosolic Ca2+ levels, hypercontracture of muscle fibers, cell necrosis) have been confirmed in amazing detail by subsequent work in a variety of models. The explicit implication of the hypothesis was that it "may provide the basis for a more rational treatment for some conditions even before their primary causes are known" (Wrogemann and Pena, 1976, Lancet, 1, 672-674). This prediction is being fulfilled, and the potential of mitochondria as pharmacological targets in muscle diseases may soon become a reality, particularly through inhibition of the mitochondrial permeability transition pore and its regulator cyclophilin D.
Collapse
Affiliation(s)
- Alessandra Zulian
- CNR Neuroscience Institute and Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marco Schiavone
- CNR Neuroscience Institute and Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Valentina Giorgio
- CNR Neuroscience Institute and Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Paolo Bernardi
- CNR Neuroscience Institute and Department of Biomedical Sciences, University of Padova, Padova, Italy.
| |
Collapse
|