1
|
Kruglov AG, Nikiforova AB. The Switching of the Type of a ROS Signal from Mitochondria: The Role of Respiratory Substrates and Permeability Transition. Antioxidants (Basel) 2024; 13:1317. [PMID: 39594458 PMCID: PMC11591497 DOI: 10.3390/antiox13111317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Flashes of superoxide anion (O2-) in mitochondria are generated spontaneously or during the opening of the permeability transition pore (mPTP) and a sudden change in the metabolic state of a cell. Under certain conditions, O2- can leave the mitochondrial matrix and perform signaling functions beyond mitochondria. In this work, we studied the kinetics of the release of O2- and H2O2 from isolated mitochondria upon mPTP opening and the modulation of the metabolic state of mitochondria by the substrates of respiration and oxidative phosphorylation. It was found that mPTP opening leads to suppression of H2O2 emission and activation of the O2- burst. When the induction of mPTP was blocked by its antagonists (cyclosporine A, ruthenium red, EGTA), the level of substrates of respiration and oxidative phosphorylation and the selective inhibitors of complexes I and V determined the type of reactive oxygen species (ROS) emitted by mitochondria. It was concluded that upon complete and partial reduction and complete oxidation of redox centers of the respiratory chain, mitochondria emit H2O2, O2-, and nothing, respectively. The results indicate that the mPTP- and substrate-dependent switching of the type of ROS leaving mitochondria may be the basis for O2-- and H2O2-selective redox signaling in a cell.
Collapse
Affiliation(s)
- Alexey G. Kruglov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Institutskaya 3, Pushchino 142290, Moscow Region, Russia;
| | | |
Collapse
|
2
|
Li A, Yi J, Li X, Dong L, Ostrow LW, Ma J, Zhou J. Deficient Sarcolemma Repair in ALS: A Novel Mechanism with Therapeutic Potential. Cells 2022; 11:cells11203263. [PMID: 36291129 PMCID: PMC9600524 DOI: 10.3390/cells11203263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Revised: 10/07/2022] [Accepted: 10/14/2022] [Indexed: 11/17/2022] Open
Abstract
The plasma membrane (sarcolemma) of skeletal muscle myofibers is susceptible to injury caused by physical and chemical stresses during normal daily movement and/or under disease conditions. These acute plasma membrane disruptions are normally compensated by an intrinsic membrane resealing process involving interactions of multiple intracellular proteins including dysferlin, annexin, caveolin, and Mitsugumin 53 (MG53)/TRIM72. There is new evidence for compromised muscle sarcolemma repair mechanisms in Amyotrophic Lateral Sclerosis (ALS). Mitochondrial dysfunction in proximity to neuromuscular junctions (NMJs) increases oxidative stress, triggering MG53 aggregation and loss of its function. Compromised membrane repair further worsens sarcolemma fragility and amplifies oxidative stress in a vicious cycle. This article is to review existing literature supporting the concept that ALS is a disease of oxidative-stress induced disruption of muscle membrane repair that compromise the integrity of the NMJs and hence augmenting muscle membrane repair mechanisms could represent a viable therapeutic strategy for ALS.
Collapse
Affiliation(s)
- Ang Li
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Jianxun Yi
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Xuejun Li
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Li Dong
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Lyle W. Ostrow
- Department of Neurology, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19122, USA
- Correspondence: (L.W.O.); (J.M.); (J.Z.)
| | - Jianjie Ma
- Department of Surgery, University of Virginia, Charlottesville, VA 22903, USA
- Correspondence: (L.W.O.); (J.M.); (J.Z.)
| | - Jingsong Zhou
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
- Correspondence: (L.W.O.); (J.M.); (J.Z.)
| |
Collapse
|
3
|
Protasi F, Girolami B, Serano M, Pietrangelo L, Paolini C. Ablation of Calsequestrin-1, Ca 2+ unbalance, and susceptibility to heat stroke. Front Physiol 2022; 13:1033300. [PMID: 36311237 PMCID: PMC9598425 DOI: 10.3389/fphys.2022.1033300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 12/05/2022] Open
Abstract
Introduction: Ca2+ levels in adult skeletal muscle fibers are mainly controlled by excitation-contraction (EC) coupling, a mechanism that translates action potentials in release of Ca2+ from the sarcoplasmic reticulum (SR) release channels, i.e. the ryanodine receptors type-1 (RyR1). Calsequestrin (Casq) is a protein that binds large amounts of Ca2+ in the lumen of the SR terminal cisternae, near sites of Ca2+ release. There is general agreement that Casq is not only important for the SR ability to store Ca2+, but also for modulating the opening probability of the RyR Ca2+ release channels. The initial studies: About 20 years ago we generated a mouse model lacking Casq1 (Casq1-null mice), the isoform predominantly expressed in adult fast twitch skeletal muscle. While the knockout was not lethal as expected, lack of Casq1 caused a striking remodeling of membranes of SR and of transverse tubules (TTs), and mitochondrial damage. Functionally, CASQ1-knockout resulted in reduced SR Ca2+ content, smaller Ca2+ transients, and severe SR depletion during repetitive stimulation. The myopathic phenotype of Casq1-null mice: After the initial studies, we discovered that Casq1-null mice were prone to sudden death when exposed to halogenated anaesthetics, heat and even strenuous exercise. These syndromes are similar to human malignant hyperthermia susceptibility (MHS) and environmental-exertional heat stroke (HS). We learned that mechanisms underlying these syndromes involved excessive SR Ca2+ leak and excessive production of oxidative species: indeed, mortality and mitochondrial damage were significantly prevented by administration of antioxidants and reduction of oxidative stress. Though, how Casq1-null mice could survive without the most important SR Ca2+ binding protein was a puzzling issue that was not solved. Unravelling the mystery: The mystery was finally solved in 2020, when we discovered that in Casq1-null mice the SR undergoes adaptations that result in constitutively active store-operated Ca2+ entry (SOCE). SOCE is a mechanism that allows skeletal fibers to use external Ca2+ when SR stores are depleted. The post-natal compensatory mechanism that allows Casq1-null mice to survive involves the assembly of new SR-TT junctions (named Ca2+ entry units) containing Stim1 and Orai1, the two proteins that mediate SOCE.
Collapse
Affiliation(s)
- Feliciano Protasi
- Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Barbara Girolami
- Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Matteo Serano
- Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Laura Pietrangelo
- Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
- Department of Medicine and Aging Sciences, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| | - Cecilia Paolini
- Center for Advanced Studies and Technology, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
- Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti-Pescara, Chieti, Italy
| |
Collapse
|
4
|
Beaufils M, Travard L, Rendu J, Marty I. Therapies for RYR1-Related Myopathies: Where We Stand and the Perspectives. Curr Pharm Des 2021; 28:15-25. [PMID: 34514983 DOI: 10.2174/1389201022666210910102516] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 08/13/2021] [Indexed: 11/22/2022]
Abstract
RyR1-related myopathies are a family of genetic neuromuscular diseases due to mutations in the RYR1 gene. No treatment exists for any of these myopathies today, which could change in the coming years with the growing number of studies dedicated to the pre-clinical assessment of various approaches, from pharmacological to gene therapy strategies, using the numerous models developed up to now. In addition, the first clinical trials for these rare diseases have just been completed or are being launched. We review the most recent results obtained for the treatment of RyR1-related myopathies, and, in view of the progress in therapeutic development for other myopathies, we discuss the possible future therapeutic perspectives for RyR1-related myopathies.
Collapse
Affiliation(s)
- Mathilde Beaufils
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble. France
| | - Lauriane Travard
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble. France
| | - John Rendu
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble. France
| | - Isabelle Marty
- University Grenoble Alpes, INSERM, U1216, CHU Grenoble Alpes, Grenoble Institut Neurosciences, Grenoble. France
| |
Collapse
|
5
|
|
6
|
Li A, Li X, Yi J, Ma J, Zhou J. Butyrate Feeding Reverses CypD-Related Mitoflash Phenotypes in Mouse Myofibers. Int J Mol Sci 2021; 22:7412. [PMID: 34299032 PMCID: PMC8304904 DOI: 10.3390/ijms22147412] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/05/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Mitoflashes are spontaneous transients of the biosensor mt-cpYFP. In cardiomyocytes, mitoflashes are associated with the cyclophilin D (CypD) mediated opening of mitochondrial permeability transition pore (mPTP), while in skeletal muscle they are considered hallmarks of mitochondrial respiration burst under physiological conditions. Here, we evaluated the potential association between mitoflashes and the mPTP opening at different CypD levels and phosphorylation status by generating three CypD derived fusion constructs with a red shifted, pH stable Ca2+ sensor jRCaMP1b. We observed perinuclear mitochondrial Ca2+ efflux accompanying mitoflashes in CypD and CypDS42A (a phosphor-resistant mutation at Serine 42) overexpressed myofibers but not the control myofibers expressing the mitochondria-targeting sequence of CypD (CypDN30). Assisted by a newly developed analysis program, we identified shorter, more frequent mitoflash activities occurring over larger areas in CypD and CypDS42A overexpressed myofibers than the control CypDN30 myofibers. These observations provide an association between the elevated CypD expression and increased mitoflash activities in hindlimb muscles in an amyotrophic lateral sclerosis (ALS) mouse model previously observed. More importantly, feeding the mice with sodium butyrate reversed the CypD-associated mitoflash phenotypes and protected against ectopic upregulation of CypD, unveiling a novel molecular mechanism underlying butyrate mediated alleviation of ALS progression in the mouse model.
Collapse
Affiliation(s)
- Ang Li
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76010, USA; (X.L.); (J.Y.)
| | - Xuejun Li
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76010, USA; (X.L.); (J.Y.)
| | - Jianxun Yi
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76010, USA; (X.L.); (J.Y.)
| | - Jianjie Ma
- Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA;
| | - Jingsong Zhou
- Department of Kinesiology, College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76010, USA; (X.L.); (J.Y.)
| |
Collapse
|
7
|
Mailloux RJ. An update on methods and approaches for interrogating mitochondrial reactive oxygen species production. Redox Biol 2021; 45:102044. [PMID: 34157640 PMCID: PMC8220584 DOI: 10.1016/j.redox.2021.102044] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/11/2021] [Indexed: 12/11/2022] Open
Abstract
The chief ROS formed by mitochondria are superoxide (O2·−) and hydrogen peroxide (H2O2). Superoxide is converted rapidly to H2O2 and therefore the latter is the chief ROS emitted by mitochondria into the cell. Once considered an unavoidable by-product of aerobic respiration, H2O2 is now regarded as a central mitokine used in mitochondrial redox signaling. However, it has been postulated that O2·− can also serve as a signal in mammalian cells. Progress in understanding the role of mitochondrial H2O2 in signaling is due to significant advances in the development of methods and technologies for its detection. Unfortunately, the development of techniques to selectively measure basal O2·− changes has been met with more significant hurdles due to its short half-life and the lack of specific probes. The development of sensitive techniques for the selective and real time measure of O2·− and H2O2 has come on two fronts: development of genetically encoded fluorescent proteins and small molecule reporters. In 2015, I published a detailed comprehensive review on the state of knowledge for mitochondrial ROS production and how it is controlled, which included an in-depth discussion of the up-to-date methods utilized for the detection of both superoxide (O2·−) and H2O2. In the article, I presented the challenges associated with utilizing these probes and their significance in advancing our collective understanding of ROS signaling. Since then, many other authors in the field of Redox Biology have published articles on the challenges and developments detecting O2·− and H2O2 in various organisms [[1], [2], [3]]. There has been significant advances in this state of knowledge, including the development of novel genetically encoded fluorescent H2O2 probes, several O2·− sensors, and the establishment of a toolkit of inhibitors and substrates for the interrogation of mitochondrial H2O2 production and the antioxidant defenses utilized to maintain the cellular H2O2 steady-state. Here, I provide an update on these methods and their implementation in furthering our understanding of how mitochondria serve as cell ROS stabilizing devices for H2O2 signaling. Details on the toolkit for interrogating the 12 sites for mitochondrial ROS production. Approaches to assess mitochondrial ROS clearance. Novel genetically encoded H2O2 sensors. Small chemical probes for sensitive detection of O2·−.
Collapse
Affiliation(s)
- Ryan J Mailloux
- The School of Human Nutrition, Faculty of Agricultural and Environmental Sciences, McGill University, Sainte-Anne-de-Bellevue, Canada.
| |
Collapse
|
8
|
Bojko B, Vasiljevic T, Boyaci E, Roszkowska A, Kraeva N, Ibarra Moreno CA, Koivu A, Wąsowicz M, Hanna A, Hamilton S, Riazi S, Pawliszyn J. Untargeted metabolomics profiling of skeletal muscle samples from malignant hyperthermia susceptible patients. Can J Anaesth 2021; 68:761-772. [PMID: 33403543 PMCID: PMC8185566 DOI: 10.1007/s12630-020-01895-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 07/29/2020] [Accepted: 07/29/2020] [Indexed: 01/09/2023] Open
Abstract
PURPOSE Malignant hyperthermia (MH) is a potentially fatal hypermetabolic condition triggered by certain anesthetics and caused by defective calcium homeostasis in skeletal muscle cells. Recent evidence has revealed impairment of various biochemical pathways in MH-susceptible patients in the absence of anesthetics. We hypothesized that clinical differences between MH-susceptible and control individuals are reflected in measurable differences in myoplasmic metabolites. METHODS We performed metabolomic profiling of skeletal muscle samples from MH-negative (control) individuals and MH-susceptible patients undergoing muscle biopsy for diagnosis of MH susceptibility. Cellular metabolites were extracted from 33 fresh and 87 frozen human muscle samples using solid phase microextraction and Metabolon® untargeted biochemical profiling platforms, respectively. Ultra-performance liquid chromatography-high resolution mass spectrometry was used for metabolite identification and validation, followed by analysis of differences in metabolites between the MH-susceptible and MH-negative groups. RESULTS Significant fold-change differences between the MH-susceptible and control groups in metabolites from various pathways were found (P value range: 0.009 to < 0.001). These included accumulation of long chain acylcarnitines, diacylglycerols, phosphoenolpyruvate, histidine pathway metabolites, lysophosphatidylcholine, oxidative stress markers, and phosphoinositols, as well as decreased levels of monoacylglycerols. The results from both analytical platforms were in agreement. CONCLUSION This metabolomics study indicates a shift from utilization of carbohydrates towards lipids for energy production in MH-susceptible individuals. This shift may result in inefficiency of beta-oxidation, and increased muscle protein turnover, oxidative stress, and/or lysophosphatidylcholine levels.
Collapse
Affiliation(s)
- Barbara Bojko
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada
- Department of Pharmacodynamics and Molecular Pharmacology, Faculty of Pharmacy, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Bydgoszcz, Poland
| | - Tijana Vasiljevic
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada
| | - Ezel Boyaci
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada
- Department of Chemistry, Middle East Technical University, Ankara, Turkey
| | - Anna Roszkowska
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada
- Department of Pharmaceutical Chemistry, Medical University of Gdansk, Gdansk, Poland
| | - Natalia Kraeva
- Malignant Hyperthermia Investigation Unit, Department of Anesthesia, University Health Network, University of Toronto, 323-200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Carlos A Ibarra Moreno
- Malignant Hyperthermia Investigation Unit, Department of Anesthesia, University Health Network, University of Toronto, 323-200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Annabel Koivu
- Malignant Hyperthermia Investigation Unit, Department of Anesthesia, University Health Network, University of Toronto, 323-200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Marcin Wąsowicz
- Malignant Hyperthermia Investigation Unit, Department of Anesthesia, University Health Network, University of Toronto, 323-200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada
| | - Amy Hanna
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Susan Hamilton
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Sheila Riazi
- Malignant Hyperthermia Investigation Unit, Department of Anesthesia, University Health Network, University of Toronto, 323-200 Elizabeth Street, Toronto, ON, M5G 2C4, Canada.
| | - Janusz Pawliszyn
- Department of Chemistry, University of Waterloo, Waterloo, ON, Canada
| |
Collapse
|
9
|
Gao M, Qin Y, Li A, Wei S, Liu B, Tian X, Gong G. Mitoflash generated at the Qo site of mitochondrial Complex III. J Cell Physiol 2021; 236:2920-2933. [PMID: 32930405 DOI: 10.1002/jcp.30059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 08/17/2020] [Accepted: 09/04/2020] [Indexed: 01/13/2023]
Abstract
The previous research has shown that mitochondrial flash (mitoflash) genesis are functionally and mechanistically integrated with mitochondrial electron transport chain (ETC) energy metabolism. However, the response of mitoflash to superoxide is not entirely consistent with the response of MitoSOX Red. The generation mechanism of mitoflash is still unclear. Here, we investigated mitoflash activities, using the different combinations of ETC substrates and inhibitors, in permeabilized cardiomyocytes or hearts. We found that blocking the complete electron flow, from Complex I to IV, with any one of ETC inhibitors including rotenone (Rot), antimycin A (AntA), myxothiazol (Myxo), stigmatellin, and sodium cyanide, will lead to the abolishment of mitoflashes triggered by substrates in adult permeabilized cardiomyocytes. However, Myxo boosted mitoflashes triggered by the reverse electron of N,N,N',N'-tetramethyl-p-phenylenediamine/ascorbate. Moreover, Rot and AntA furtherly enhanced mitoflash activity rather than depressed it, suggesting that mitoflashes generated at the Complex III Qo site. Meanwhile, the inhibition of Complex III protein expression resulted in the activity of Complex III decrease, which decreased mitoflash frequency. The function defect (no change of protein level) of the Qo site of Complex III in aging hearts augmented mitoflash generation confirmed the Qo site function was critical to mitoflash genesis. Thus, our results indicate that mitoflash detected by circularly permuted yellow fluorescent protein is generated at the Qo site of Complex III.
Collapse
Affiliation(s)
- Meng Gao
- Institute for Regenerative Medicine, Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yuan Qin
- Department of Pharmacy, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Anqi Li
- Institute for Regenerative Medicine, Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Sailei Wei
- Institute for Regenerative Medicine, Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Bilin Liu
- Institute for Regenerative Medicine, Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiangang Tian
- Institute for Regenerative Medicine, Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
- Department of Cardiovascular Surgery, Daping Hospital, Army Medical Center of PLA, Chongqing, China
| | - Guohua Gong
- Institute for Regenerative Medicine, Research Center for Translational Medicine, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
10
|
Van Wijk R, Van Wijk EP, Pang J, Yang M, Yan Y, Han J. Integrating Ultra-Weak Photon Emission Analysis in Mitochondrial Research. Front Physiol 2020; 11:717. [PMID: 32733265 PMCID: PMC7360823 DOI: 10.3389/fphys.2020.00717] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 05/29/2020] [Indexed: 12/11/2022] Open
Abstract
Once regarded solely as the energy source of the cell, nowadays mitochondria are recognized to perform multiple essential functions in addition to energy production. Since the discovery of pathogenic mitochondrial DNA defects in the 1980s, research advances have revealed an increasing number of common human diseases, which share an underlying pathogenesis involving mitochondrial dysfunction. A major factor in this dysfunction is reactive oxygen species (ROS), which influence the mitochondrial-nuclear crosstalk and the link with the epigenome, an influence that provides explanations for pathogenic mechanisms. Regarding these mechanisms, we should take into account that mitochondria produce the majority of ultra-weak photon emission (UPE), an aspect that is often ignored - this type of emission may serve as assay for ROS, thus providing new opportunities for a non-invasive diagnosis of mitochondrial dysfunction. In this article, we overviewed three relevant areas of mitochondria-related research over the period 1960-2020: (a) respiration and energy production, (b) respiration-related production of free radicals and other ROS species, and (c) ultra-weak photon emission in relation to ROS and stress. First, we have outlined how these research areas initially developed independently of each other - following that, our review aims to show their stepwise integration during later stages of development. It is suggested that a further stimulation of research on UPE may have the potential to enhance the progress of modern mitochondrial research and its integration in medicine.
Collapse
Affiliation(s)
- Roeland Van Wijk
- Meluna Research, Department of Biophotonics, Geldermalsen, Netherlands
| | | | - Jingxiang Pang
- Key Laboratory for Biotech-Drugs of National Health Commission, Shandong Medicinal Biotechnology Center, Jinan, China
- Shandong First Medical University, Jinan, China
- Shandong Academy of Medical Sciences, Jinan, China
| | - Meina Yang
- Key Laboratory for Biotech-Drugs of National Health Commission, Shandong Medicinal Biotechnology Center, Jinan, China
- Shandong First Medical University, Jinan, China
- Shandong Academy of Medical Sciences, Jinan, China
| | - Yu Yan
- Meluna Research, Department of Biophotonics, Geldermalsen, Netherlands
| | - Jinxiang Han
- Key Laboratory for Biotech-Drugs of National Health Commission, Shandong Medicinal Biotechnology Center, Jinan, China
- Shandong First Medical University, Jinan, China
- Shandong Academy of Medical Sciences, Jinan, China
| |
Collapse
|
11
|
Nikolaidis MG, Margaritelis NV, Matsakas A. Quantitative Redox Biology of Exercise. Int J Sports Med 2020; 41:633-645. [PMID: 32455453 DOI: 10.1055/a-1157-9043] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Biology is rich in claims that reactive oxygen and nitrogen species are involved in every biological process and disease. However, many quantitative aspects of redox biology remain elusive. The important quantitative parameters you need to address the feasibility of redox reactions in vivo are: rate of formation and consumption of a reactive oxygen and nitrogen species, half-life, diffusibility and membrane permeability. In the first part, we explain the basic chemical kinetics concepts and algebraic equations required to perform "street fighting" quantitative analysis. In the second part, we provide key numbers to help thinking about sizes, concentrations, rates and other important quantities that describe the major oxidants (superoxide, hydrogen peroxide, nitric oxide) and antioxidants (vitamin C, vitamin E, glutathione). In the third part, we present the quantitative effect of exercise on superoxide, hydrogen peroxide and nitric oxide concentration in mitochondria and whole muscle and calculate how much hydrogen peroxide concentration needs to increase to transduce signalling. By taking into consideration the quantitative aspects of redox biology we can: i) refine the broad understanding of this research area, ii) design better future studies and facilitate comparisons among studies, and iii) define more efficiently the "borders" between cellular signaling and stress.
Collapse
Affiliation(s)
- Michalis G Nikolaidis
- Department of Physical Education and Sport Sciences at Serres, Aristotle University of Thessaloniki, Serres, Greece
| | - Nikos V Margaritelis
- Department of Physical Education and Sport Sciences at Serres, Aristotle University of Thessaloniki, Serres, Greece.,General Military Hospital of Thessaloniki, Dialysis Unit, Thessaloniki, Greece
| | - Antonios Matsakas
- Centre for Atherothrombotic & Metabolic Disease, Hull York Medical School, Hull, United Kingdom of Great Britain and Northern Ireland
| |
Collapse
|
12
|
Lawal TA, Wires ES, Terry NL, Dowling JJ, Todd JJ. Preclinical model systems of ryanodine receptor 1-related myopathies and malignant hyperthermia: a comprehensive scoping review of works published 1990-2019. Orphanet J Rare Dis 2020; 15:113. [PMID: 32381029 PMCID: PMC7204063 DOI: 10.1186/s13023-020-01384-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Pathogenic variations in the gene encoding the skeletal muscle ryanodine receptor (RyR1) are associated with malignant hyperthermia (MH) susceptibility, a life-threatening hypermetabolic condition and RYR1-related myopathies (RYR1-RM), a spectrum of rare neuromuscular disorders. In RYR1-RM, intracellular calcium dysregulation, post-translational modifications, and decreased protein expression lead to a heterogenous clinical presentation including proximal muscle weakness, contractures, scoliosis, respiratory insufficiency, and ophthalmoplegia. Preclinical model systems of RYR1-RM and MH have been developed to better understand underlying pathomechanisms and test potential therapeutics. METHODS We conducted a comprehensive scoping review of scientific literature pertaining to RYR1-RM and MH preclinical model systems in accordance with the PRISMA Scoping Reviews Checklist and the framework proposed by Arksey and O'Malley. Two major electronic databases (PubMed and EMBASE) were searched without language restriction for articles and abstracts published between January 1, 1990 and July 3, 2019. RESULTS Our search yielded 5049 publications from which 262 were included in this review. A majority of variants tested in RYR1 preclinical models were localized to established MH/central core disease (MH/CCD) hot spots. A total of 250 unique RYR1 variations were reported in human/rodent/porcine models with 95% being missense substitutions. The most frequently reported RYR1 variant was R614C/R615C (human/porcine total n = 39), followed by Y523S/Y524S (rabbit/mouse total n = 30), I4898T/I4897T/I4895T (human/rabbit/mouse total n = 20), and R163C/R165C (human/mouse total n = 18). The dyspedic mouse was utilized by 47% of publications in the rodent category and its RyR1-null (1B5) myotubes were transfected in 23% of publications in the cellular model category. In studies of transfected HEK-293 cells, 57% of RYR1 variations affected the RyR1 channel and activation core domain. A total of 15 RYR1 mutant mouse strains were identified of which ten were heterozygous, three were compound heterozygous, and a further two were knockout. Porcine, avian, zebrafish, C. elegans, canine, equine, and drosophila model systems were also reported. CONCLUSIONS Over the past 30 years, there were 262 publications on MH and RYR1-RM preclinical model systems featuring more than 200 unique RYR1 variations tested in a broad range of species. Findings from these studies have set the foundation for therapeutic development for MH and RYR1-RM.
Collapse
Affiliation(s)
- Tokunbor A Lawal
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Emily S Wires
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Nancy L Terry
- National Institutes of Health Library, National Institutes of Health, Bethesda, MD, USA
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joshua J Todd
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
13
|
Lavorato M, Formenti F, Franzini-Armstrong C. The structural basis for intermitochondrial communications is fundamentally different in cardiac and skeletal muscle. Exp Physiol 2020; 105:606-612. [PMID: 32189419 DOI: 10.1113/ep087503] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 05/22/2019] [Indexed: 01/15/2023]
Abstract
NEW FINDINGS What is the topic for this review? This review summarizes recent discoveries in mitochondrial development and morphology studied with electron microscopy. What advances does it highlight? Although mitochondria are generally considered to be isolated from each other, this review highlights recently discovered evidence for the presence of intermitochondrial communication structures in cardiac and skeletal muscle, in animal models and humans. Within striated muscles, the means of intermitochondrial exchange and the reaction of mitochondria to external stimuli are uniquely dependent on the tissue, and we clearly differentiate between nanotunnels, the active protrusion of cardiac mitochondria, and the connecting ducts of skeletal muscle derived from fusion-fission and elongation events. ABSTRACT This review focuses on recent discoveries in skeletal and cardiac muscles indicating that mitochondria behave as an interactive cohort with inter-organelle communication and specific reactions to stress signals. Our new finding is that intermitochondrial communications in cardiac and skeletal muscles rely on two distinct methods. In cardiac muscle, mitochondria are discrete entities and are fairly well immobilized in a structural context. The organelles have developed a unique method of communication, via nanotunnels, which allow temporary connection from one mitochondrion to another over distances of up to several micrometres, without overall movement of the individual organelles and loss of their identity. Skeletal muscle mitochondria, in contrast, are dynamic. Through fusion, fission and elongation, they form connections that include constrictions and connecting ducts (distinct from nanotunnels) and lose individual identity in the formation of extensive networks. Connecting elements in skeletal muscle are distinct from nanotunnels in cardiac muscle.
Collapse
Affiliation(s)
- Manuela Lavorato
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA, USA.,Department of Genetics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Federico Formenti
- Centre for Human and Applied Physiological Sciences, King's College London, London, UK.,Nuffield Division of Anaesthetics, University of Oxford, Oxford, UK.,Department of Biomechanics, University of Nebraska at Omaha, Omaha, NE, USA
| | | |
Collapse
|
14
|
McMurray F, MacFarlane M, Kim K, Patten DA, Wei-LaPierre L, Fullerton MD, Harper ME. Maternal diet–induced obesity alters muscle mitochondrial function in offspring without changing insulin sensitivity. FASEB J 2019; 33:13515-13526. [DOI: 10.1096/fj.201901150r] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Fiona McMurray
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| | - Megan MacFarlane
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Kijoo Kim
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David A. Patten
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| | - Lan Wei-LaPierre
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, New York, USA
| | - Morgan D. Fullerton
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
- Ottawa Institute of Systems Biology, Ottawa, Ontario, Canada
| |
Collapse
|
15
|
Canato M, Capitanio P, Cancellara L, Leanza L, Raffaello A, Reane DV, Marcucci L, Michelucci A, Protasi F, Reggiani C. Excessive Accumulation of Ca 2 + in Mitochondria of Y522S-RYR1 Knock-in Mice: A Link Between Leak From the Sarcoplasmic Reticulum and Altered Redox State. Front Physiol 2019; 10:1142. [PMID: 31607937 PMCID: PMC6755340 DOI: 10.3389/fphys.2019.01142] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 08/21/2019] [Indexed: 12/04/2022] Open
Abstract
Mice (Y522S or YS), carrying a mutation of the sarcoplasmic reticulum (SR) Ca2+ release channel of skeletal muscle fibers (ryanodine receptor type-1, RyR1) which causes Ca2+ leak, are a widely accepted and intensively studied model for human malignant hyperthermia (MH) susceptibility. Since the involvement of reactive oxygen species (ROS) and of mitochondria in MH crisis has been previously debated, here we sought to determine Ca2+ uptake in mitochondria and its possible link with ROS production in single fibers isolated from flexor digitorum brevis (FDB) of YS mice. We found that Ca2+ concentration in the mitochondrial matrix, as detected with the ratiometric FRET-based 4mtD3cpv probe, was higher in YS than in wild-type (WT) fibers at rest and after Ca2+ release from SR during repetitive electrical stimulation or caffeine administration. Also mitochondrial ROS production associated with contractile activity (detected with Mitosox probe) was much higher in YS fibers than in WT. Importantly, the inhibition of mitochondrial Ca2+ uptake achieved by silencing MCU reduced ROS accumulation in the matrix and Ca2+ release from SR. Finally, inhibition of mitochondrial ROS accumulation using Mitotempo reduced SR Ca2+ release in YS fibers exposed to caffeine. The present results support the view that mitochondria take up larger amounts of Ca2+ in YS than in WT fibers and that mitochondrial ROS production substantially contributes to the increased caffeine-sensitivity and to the enhanced Ca2+ release from SR in YS fibers.
Collapse
Affiliation(s)
- Marta Canato
- Department of Biomedical Sciences, School of Medicine and Surgery, University of Padova, Padua, Italy
| | - Paola Capitanio
- Department of Biomedical Sciences, School of Medicine and Surgery, University of Padova, Padua, Italy
| | - Lina Cancellara
- Department of Biomedical Sciences, School of Medicine and Surgery, University of Padova, Padua, Italy
| | - Luigi Leanza
- Department of Biology, University of Padova, Padua, Italy
| | - Anna Raffaello
- Department of Biomedical Sciences, School of Medicine and Surgery, University of Padova, Padua, Italy
| | - Denis Vecellio Reane
- Department of Biomedical Sciences, School of Medicine and Surgery, University of Padova, Padua, Italy
| | - Lorenzo Marcucci
- Department of Biomedical Sciences, School of Medicine and Surgery, University of Padova, Padua, Italy
| | - Antonio Michelucci
- Center for Advanced Studies and Technology, Università degli Studi “G. d’Annunzio” Chieti–Pescara, Chieti, Italy
- Department of Medicine and Aging Sciences, Università degli Studi “G. d’Annunzio” Chieti–Pescara, Chieti, Italy
| | - Feliciano Protasi
- Center for Advanced Studies and Technology, Università degli Studi “G. d’Annunzio” Chieti–Pescara, Chieti, Italy
- Department of Medicine and Aging Sciences, Università degli Studi “G. d’Annunzio” Chieti–Pescara, Chieti, Italy
| | - Carlo Reggiani
- Department of Biomedical Sciences, School of Medicine and Surgery, University of Padova, Padua, Italy
- Institute for Kinesiology Research, Science and Research Center of Koper, Koper, Slovenia
| |
Collapse
|
16
|
Wei-LaPierre L, Dirksen RT. Isolating a reverse-mode ATP synthase-dependent mechanism of mitoflash activation. J Gen Physiol 2019; 151:708-713. [PMID: 31010808 PMCID: PMC6571996 DOI: 10.1085/jgp.201912358] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Wei-LaPierre and Dirksen discuss new work investigating the molecular events underlying mitoflash biogenesis.
Collapse
Affiliation(s)
- Lan Wei-LaPierre
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY
| |
Collapse
|
17
|
Wei-LaPierre L, Ainbinder A, Tylock KM, Dirksen RT. Substrate-dependent and cyclophilin D-independent regulation of mitochondrial flashes in skeletal and cardiac muscle. Arch Biochem Biophys 2019; 665:122-131. [PMID: 30872061 PMCID: PMC6499064 DOI: 10.1016/j.abb.2019.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 03/05/2019] [Accepted: 03/06/2019] [Indexed: 01/20/2023]
Abstract
Mitochondrial flashes (mitoflashes) are stochastic events in the mitochondrial matrix detected by mitochondrial-targeted cpYFP (mt-cpYFP). Mitoflashes are quantal bursts of reactive oxygen species (ROS) production accompanied by modest matrix alkalinization and depolarization of the mitochondrial membrane potential. Mitoflashes are fundamental events present in a wide range of cell types. To date, the precise mechanisms for mitoflash generation and termination remain elusive. Transient opening of the mitochondrial membrane permeability transition pore (mPTP) during a mitoflash is proposed to account for the mitochondrial membrane potential depolarization. Here, we set out to compare the tissue-specific effects of cyclophilin D (CypD)-deficiency and mitochondrial substrates on mitoflash activity in skeletal and cardiac muscle. In contrast to previous reports, we found that CypD knockout did not alter the mitoflash frequency or other mitoflash properties in acutely isolated cardiac myocytes, skeletal muscle fibers, or isolated mitochondria from skeletal muscle and the heart. However, in skeletal muscle fibers, CypD deficiency resulted in a parallel increase in both activity-dependent mitochondrial Ca2+ uptake and activity-dependent mitoflash activity. Increases in both mitochondrial Ca2+ uptake and mitoflash activity following electrical stimulation were abolished by inhibition of mitochondrial Ca2+ uptake. We also found that mitoflash frequency and amplitude differ greatly between intact skeletal muscle fibers and cardiac myocytes, but that this difference is absent in isolated mitochondria. We propose that this difference may be due, in part, to differences in substrate availability in intact skeletal muscle fibers (primarily glycolytic) and cardiac myocytes (largely oxidative). Overall, we find that CypD does not contribute significantly in mitoflash biogenesis under basal conditions in skeletal and cardiac muscle, but does regulate mitoflash events during muscle activity. In addition, tissue-dependent differences in mitoflash frequency are strongly regulated by mitochondrial substrate availability.
Collapse
Affiliation(s)
- Lan Wei-LaPierre
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA.
| | - Alina Ainbinder
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Kevin M Tylock
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY, 14642, USA
| |
Collapse
|
18
|
Zhang C, Sun F, Xiong B, Zhang Z. Preparation of mitochondria to measure superoxide flashes in angiosperm flowers. PeerJ 2019; 7:e6708. [PMID: 30997289 PMCID: PMC6462395 DOI: 10.7717/peerj.6708] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2018] [Accepted: 03/02/2019] [Indexed: 11/20/2022] Open
Abstract
Background Mitochondria are the center of energy metabolism and the production of reactive oxygen species (ROS). ROS production results in a burst of “superoxide flashes”, which is always accompanied by depolarization of mitochondrial membrane potential. Superoxide flashes have only been studied in the model plant Arabidopsis thaliana using a complex method to isolate mitochondria. In this study, we present an efficient, easier method to isolate functional mitochondria from floral tissues to measure superoxide flashes. Method We used 0.5 g samples to isolate mitochondria within <1.5 h from flowers of two non-transgenic plants (Magnolia denudata and Nelumbo nucifera) to measure superoxide flashes. Superoxide flashes were visualized by the pH-insensitive indicator MitoSOX Red, while the mitochondrial membrane potential (ΔΨ m) was labelled with TMRM. Results Mitochondria isolated using our method showed a high respiration ratio. Our results indicate that the location of ROS and mitochondria was in a good coincidence. Increased ROS together with a higher frequency of superoxide flashes was found in mitochondria isolated from the flower pistil. Furthermore, a higher rate of depolarization of the ΔΨ m was observed in the pistil. Taken together, these results demonstrate that the frequency of superoxide flashes is closely related to depolarization of the ΔΨ m in petals and pistils of flowers.
Collapse
Affiliation(s)
- Chulan Zhang
- College of Nature Conservation, Beijing Forestry University, Beijing, China
| | - Fengshuo Sun
- College of Biological Sciences and Biotechnology, Beijing Forestry University, Beijing, China
| | - Biao Xiong
- College of Tea Science, Guizhou University, Guizhou Province, China
| | - Zhixiang Zhang
- College of Nature Conservation, Beijing Forestry University, Beijing, China
| |
Collapse
|
19
|
Feng G, Liu B, Li J, Cheng T, Huang Z, Wang X, Cheng HP. Mitoflash biogenesis and its role in the autoregulation of mitochondrial proton electrochemical potential. J Gen Physiol 2019; 151:727-737. [PMID: 30877142 PMCID: PMC6571995 DOI: 10.1085/jgp.201812176] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 11/29/2018] [Accepted: 02/19/2019] [Indexed: 01/11/2023] Open
Abstract
Individual mitochondria undergo an intermittent, all-or-none electrochemical excitation termed “mitoflash.” Feng et al. show that mitoflash occurs following build-up of mitochondrial electrochemical potential and may serve to autoregulate mitochondrial proton electrochemical potential. Respiring mitochondria undergo an intermittent electrical and chemical excitation called mitochondrial flash (mitoflash), which transiently uncouples mitochondrial respiration from ATP production. How a mitoflash is generated and what specific role it plays in bioenergetics remain incompletely understood. Here, we investigate mitoflash biogenesis in isolated cardiac mitochondria by varying the respiratory states and substrate supply and by dissecting the involvement of different electron transfer chain (ETC) complexes. We find that robust mitoflash activity occurs once mitochondria are electrochemically charged by state II/IV respiration (i.e., no ATP synthesis at Complex V), regardless of the substrate entry site (Complex I, Complex II, or Complex IV). Inhibiting forward electron transfer abolishes, while blocking reverse electron transfer generally augments, mitoflash production. Switching from state II/IV to state III respiration, to allow for ATP synthesis at Complex V, markedly diminishes mitoflash activity. Intriguingly, when mitochondria are electrochemically charged by the ATPase activity of Complex V, mitoflashes are generated independently of ETC activity. These findings suggest that mitoflash biogenesis is mechanistically linked to the build up of mitochondrial electrochemical potential rather than ETC activity alone, and may functionally counteract overcharging of the mitochondria and hence serve as an autoregulator of mitochondrial proton electrochemical potential.
Collapse
Affiliation(s)
- Gaomin Feng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Beibei Liu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Jinghang Li
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Tianlei Cheng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhanglong Huang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xianhua Wang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Heping Peace Cheng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
20
|
Zhou J, Li A, Li X, Yi J. Dysregulated mitochondrial Ca 2+ and ROS signaling in skeletal muscle of ALS mouse model. Arch Biochem Biophys 2019; 663:249-258. [PMID: 30682329 PMCID: PMC6506190 DOI: 10.1016/j.abb.2019.01.024] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 12/24/2018] [Accepted: 01/18/2019] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a devastating neuromuscular disease characterized by motor neuron loss and prominent skeletal muscle wasting. Despite more than one hundred years of research efforts, the pathogenic mechanisms underlying neuromuscular degeneration in ALS remain elusive. While the death of motor neuron is a defining hallmark of ALS, accumulated evidences suggested that in addition to being a victim of motor neuron axonal withdrawal, the intrinsic skeletal muscle degeneration may also actively contribute to ALS disease pathogenesis and progression. Examination of spinal cord and muscle autopsy/biopsy samples of ALS patients revealed similar mitochondrial abnormalities in morphology, quantity and disposition, which are accompanied by defective mitochondrial respiratory chain complex and elevated oxidative stress. Detailing the molecular/cellular mechanisms and the role of mitochondrial dysfunction in ALS relies on ALS animal model studies. This review article discusses the dysregulated mitochondrial Ca2+ and reactive oxygen species (ROS) signaling revealed in live skeletal muscle derived from ALS mouse models, and a potential role of the vicious cycle formed between the dysregulated mitochondrial Ca2+ signaling and excessive ROS production in promoting muscle wasting during ALS progression.
Collapse
Affiliation(s)
- Jingsong Zhou
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA.
| | - Ang Li
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Xuejun Li
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA
| | - Jianxun Yi
- Kansas City University of Medicine and Bioscience, Kansas City, MO 64106, USA; College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX 76019, USA.
| |
Collapse
|
21
|
Pereira YCL, Nascimento GC, Iyomasa DM, Fernández RAR, Calzzani RA, Leite-Panissi CRA, Novaes PD, Iyomasa MM. Exodontia-induced muscular hypofunction by itself or associated to chronic stress impairs masseter muscle morphology and its mitochondrial function. Microsc Res Tech 2019; 82:530-537. [PMID: 30741445 DOI: 10.1002/jemt.23196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Revised: 10/12/2018] [Accepted: 11/07/2018] [Indexed: 12/12/2022]
Abstract
Stress is associated with orofacial pain sensitivity and is qualified as a temporomandibular disorder risk factor. During stressful periods, painful thresholds of masticatory muscles in individuals suffering muscle facial pain are significantly lower than in controls, but the exact physiologic mechanism underlying this relation remains unclear. Our hypothesis is that chronic unpredictable stress and masticatory hypofunction induce morphologic and metabolic masseter muscle changes in rats. For test this hypothesis, adult Wistar rats were submitted to chronic unpredictable stress and/or exodontia of left molars and the left masseter muscle was removed for analysis. The parameters evaluated included ultrastructure, oxidative level, metabolism activity and morphological analysis in this muscle. Our data show by histological analysis, that stress and exodontia promoted a variation on diameters and also angled contours in masseter fibers. The masticatory hypofunction increased oxidative metabolism as well as decreased reactive species of oxygen in masseter muscle. The ultrastructural analysis of muscle fibers showed disruption of the sarcoplasmic reticulum cisterns in certain regions of the fiber in stress group, and the disappearance of the sarcoplasmic reticulum membrane in group with association of stress and exodontia. Our findings clarify mechanisms by which chronic stress and masticatory hypofunction might be involved in the pathophysiology of muscular dysfunctions. Masticatory hypofunction influenced oxidative stress and induced oxidative metabolism on masseter muscle, as well as altered its fiber morphology. Chronic stress presented malefic effect on masseter morphology at micro and ultra structurally. When both stimuli were applied, there were atrophic fibers and a complete mitochondrial derangement.
Collapse
Affiliation(s)
| | - Glauce Crivelaro Nascimento
- Department of Morphology, Physiology and Basic Pathology of Dentistry School of RibeirãoPreto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Daniela Mizusaki Iyomasa
- Department of Morphology, Physiology and Basic Pathology of Dentistry School of RibeirãoPreto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Rodrigo Alberto Restrepo Fernández
- Department of Morphology, Physiology and Basic Pathology of Dentistry School of RibeirãoPreto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Ricardo Alexandre Calzzani
- Department of Morphology, Physiology and Basic Pathology of Dentistry School of RibeirãoPreto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Christie Ramos Andrade Leite-Panissi
- Department of Morphology, Physiology and Basic Pathology of Dentistry School of RibeirãoPreto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.,Psychobiology Graduate Program, School of Philosophy, Science and Literature of RibeirãoPreto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| | - Pedro Duarte Novaes
- Faculty of Dentistry of Piracicaba, University of Campinas, Campinas, São Paulo, Brazil
| | - Mamie Mizusaki Iyomasa
- Department of Morphology, Physiology and Basic Pathology of Dentistry School of RibeirãoPreto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
22
|
McBride S, Wei-LaPierre L, McMurray F, MacFarlane M, Qiu X, Patten DA, Dirksen RT, Harper ME. Skeletal muscle mitoflashes, pH, and the role of uncoupling protein-3. Arch Biochem Biophys 2019; 663:239-248. [PMID: 30659802 DOI: 10.1016/j.abb.2019.01.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 12/28/2018] [Accepted: 01/15/2019] [Indexed: 01/03/2023]
Abstract
Mitochondrial reactive oxygen species (ROS) are important cellular signaling molecules, but can cause oxidative damage if not kept within tolerable limits. An important proximal form of ROS in mitochondria is superoxide. Its production is thought to occur in regulated stochastic bursts, but current methods using mitochondrial targeted cpYFP to assess superoxide flashes are confounded by changes in pH. Accordingly, these flashes are generally referred to as 'mitoflashes'. Here we provide regulatory insights into mitoflashes and pH fluctuations in skeletal muscle, and the role of uncoupling protein-3 (UCP3). Using quantitative confocal microscopy of mitoflashes in intact muscle fibers, we show that the mitoflash magnitude significantly correlates with the degree of mitochondrial inner membrane depolarization and ablation of UCP3 did not affect this correlation. We assessed the effects of the absence of UCP3 on mitoflash activity in intact skeletal muscle fibers, and found no effects on mitoflash frequency, amplitude or duration, with a slight reduction in the average size of mitoflashes. We further investigated the regulation of pH flashes (pHlashes, presumably a component of mitoflash) by UCP3 using mitochondrial targeted SypHer (mt-SypHer) in skeletal muscle fibers. The frequency of pHlashes was significantly reduced in the absence of UCP3, without changes in other flash properties. ROS scavenger, tiron, did not alter pHlash frequency in either WT or UCP3KO mice. High resolution respirometry revealed that in the absence of UCP3 there is impaired proton leak and Complex I-driven respiration and maximal coupled respiration. Total cellular production of hydrogen peroxide (H2O2) as detected by Amplex-UltraRed was unaffected. Altogether, we demonstrate a correlation between mitochondrial membrane potential and mitoflash magnitude in skeletal muscle fibers that is independent of UCP3, and a role for UCP3 in the control of pHlash frequency and of proton leak- and Complex I coupled-respiration in skeletal muscle fibers. The differential regulation of mitoflashes and pHlashes by UCP3 and tiron also indicate that the two events, though may be related, are not identical events.
Collapse
Affiliation(s)
- S McBride
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd., Ottawa, ON, K1H 8M5, Canada
| | - L Wei-LaPierre
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY, 14642-8711, USA
| | - F McMurray
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd., Ottawa, ON, K1H 8M5, Canada
| | - M MacFarlane
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd., Ottawa, ON, K1H 8M5, Canada
| | - X Qiu
- Department of Biostatistics, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY, 14642-8711, USA
| | - D A Patten
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd., Ottawa, ON, K1H 8M5, Canada
| | - R T Dirksen
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, 601 Elmwood Avenue, Rochester, NY, 14642-8711, USA
| | - M-E Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, 451 Smyth Rd., Ottawa, ON, K1H 8M5, Canada.
| |
Collapse
|
23
|
Lavorato M, Loro E, Debattisti V, Khurana TS, Franzini-Armstrong C. Elongated mitochondrial constrictions and fission in muscle fatigue. J Cell Sci 2018; 131:jcs221028. [PMID: 30404834 PMCID: PMC6288074 DOI: 10.1242/jcs.221028] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/30/2018] [Indexed: 12/24/2022] Open
Abstract
Mitochondria respond to stress and undergo fusion and fission at variable rates, depending on cell status. To understand mitochondrial behavior during muscle fatigue, we investigated mitochondrial ultrastructure and expression levels of a fission- and stress-related protein in fast-twitch muscle fibers of mice subjected to fatigue testing. Mice were subjected to running at increasing speed until exhaustion at 45 min-1 h. In further experiments, high-intensity muscle stimulation through the sciatic nerve simulated the forced treadmill exercise. We detected a rare phenotype characterized by elongated mitochondrial constrictions (EMCs) connecting two separate segments of the original organelles. EMCs are rare in resting muscles and their frequency increases, albeit still at low levels, in stimulated muscles. The constrictions are accompanied by elevated phosphorylation of Drp1 (Dnm1l) at Ser 616, indicating an increased translocation of Drp1 to the mitochondrial membrane. This is indicative of a mitochondrial stress response, perhaps leading to or facilitating a long-lasting fission event. A close apposition of sarcoplasmic reticulum (SR) to the constricted areas, detected using both transmission and scanning electron microscopy, is highly suggestive of SR involvement in inducing mitochondrial constrictions.
Collapse
Affiliation(s)
- Manuela Lavorato
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Genetics, Children's Hospital of Philadelphia, PA 19104, USA
| | - Emanuele Loro
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Valentina Debattisti
- MitoCare Center, Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Tejvir S Khurana
- Department of Physiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Clara Franzini-Armstrong
- Department of Cell and Developmental Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
24
|
Xiao Y, Karam C, Yi J, Zhang L, Li X, Yoon D, Wang H, Dhakal K, Ramlow P, Yu T, Mo Z, Ma J, Zhou J. ROS-related mitochondrial dysfunction in skeletal muscle of an ALS mouse model during the disease progression. Pharmacol Res 2018; 138:25-36. [PMID: 30236524 PMCID: PMC6263743 DOI: 10.1016/j.phrs.2018.09.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Revised: 09/07/2018] [Accepted: 09/07/2018] [Indexed: 02/06/2023]
Abstract
In amyotrophic lateral sclerosis (ALS), mitochondrial dysfunction and oxidative stress form a vicious cycle that promotes neurodegeneration and muscle wasting. To quantify the disease-stage-dependent changes of mitochondrial function and their relationship to the generation of reactive oxygen species (ROS), we generated double transgenic mice (G93A/cpYFP) that carry human ALS mutation SOD1G93A and mt-cpYFP transgenes, in which mt-cpYFP detects dynamic changes of ROS-related mitoflash events at individual mitochondria level. Compared with wild type mice, mitoflash activity in the SOD1G93A (G93A) mouse muscle showed an increased flashing frequency prior to the onset of ALS symptom (at the age of 2 months), whereas the onset of ALS symptoms (at the age of 4 months) is associated with drastic changes in the kinetics property of mitoflash signal with prolonged full duration at half maximum (FDHM). Elevated levels of cytosolic ROS in skeletal muscle derived from the SOD1G93A mice were confirmed with fluorescent probes, MitoSOX™ Red and ROS Brite™570. Immunoblotting analysis of subcellular mitochondrial fractionation of G93A muscle revealed an increased expression level of cyclophilin D (CypD), a regulatory component of the mitochondrial permeability transition pore (mPTP), at the age of 4 months but not at the age of 2 months. Transient overexpressing of SOD1G93A in skeletal muscle of wild type mice directly promoted mitochondrial ROS production with an enhanced mitoflash activity in the absence of motor neuron axonal withdrawal. Remarkably, the SOD1G93A-induced mitoflash activity was attenuated by the application of cyclosporine A (CsA), an inhibitor of CypD. Similar to the observation with the SOD1G93A transgenic mice, an increased expression level of CypD was also detected in skeletal muscle following transient overexpression of SOD1G93A. Overall, this study reveals a disease-stage-dependent change in mitochondrial function that is associated with CypD-dependent mPTP opening; and the ALS mutation SOD1G93A directly contributes to mitochondrial dysfunction in the absence of motor neuron axonal withdrawal.
Collapse
Affiliation(s)
- Yajuan Xiao
- Kansas City University of Medicine and Bioscience, Kansas City, USA; Rush University School of Medicine, Chicago, IL, USA
| | - Chehade Karam
- Rush University School of Medicine, Chicago, IL, USA
| | - Jianxun Yi
- Kansas City University of Medicine and Bioscience, Kansas City, USA; Rush University School of Medicine, Chicago, IL, USA
| | - Lin Zhang
- Kansas City University of Medicine and Bioscience, Kansas City, USA; Zunyi Medical College, Zunyi, China
| | - Xuejun Li
- Kansas City University of Medicine and Bioscience, Kansas City, USA
| | - Dosuk Yoon
- Kansas City University of Medicine and Bioscience, Kansas City, USA
| | - Huan Wang
- Kansas City University of Medicine and Bioscience, Kansas City, USA
| | - Kamal Dhakal
- Kansas City University of Medicine and Bioscience, Kansas City, USA
| | - Paul Ramlow
- Kansas City University of Medicine and Bioscience, Kansas City, USA
| | - Tian Yu
- Zunyi Medical College, Zunyi, China
| | - Zhaohui Mo
- 3rd Xiangya Hospital, Central South University, Changsha, China
| | - Jianjie Ma
- Wexner Medical Center, The Ohio State University, Columbus, OH, USA
| | - Jingsong Zhou
- Kansas City University of Medicine and Bioscience, Kansas City, USA; Rush University School of Medicine, Chicago, IL, USA.
| |
Collapse
|
25
|
Rosselin M, Nunes-Hasler P, Demaurex N. Ultrastructural Characterization of Flashing Mitochondria. ACTA ACUST UNITED AC 2018; 1:1-14. [PMID: 30406212 PMCID: PMC6217927 DOI: 10.1177/2515256418801423] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Mitochondria undergo spontaneous transient elevations in matrix pH associated with drops in mitochondrial membrane potential. These mitopHlashes require a functional respiratory chain and the profusion protein optic atrophy 1, but their mechanistic basis is unclear. To gain insight on the origin of these dynamic events, we resolved the ultrastructure of flashing mitochondria by correlative light and electron microscopy. HeLa cells expressing the matrix-targeted pH probe mitoSypHer were screened for mitopHlashes and fixed immediately after the occurrence of a flashing event. The cells were then processed for imaging by serial block face scanning electron microscopy using a focused ion beam to generate ~1,200 slices of 10 nm thickness from a 28 μm × 15 μm cellular volume. Correlation of live/fixed fluorescence and electron microscopy images allowed the unambiguous identification of flashing and nonflashing mitochondria. Three-dimensional reconstruction and surface mapping revealed that each tomogram contained two flashing mitochondria of unequal sizes, one being much larger than the average mitochondrial volume. Flashing mitochondria were 10-fold larger than silent mitochondria but with a surface to volume ratio and a cristae volume similar to nonflashing mitochondria. Flashing mitochondria were connected by tubular structures, formed more membrane contact sites, and a constriction was observed at a junction between a flashing mitochondrion and a nonflashing mitochondrion. These data indicate that flashing mitochondria are structurally preserved and bioenergetically competent but form numerous membrane contact sites and are connected by tubular structures, consistent with our earlier suggestion that mitopHlashes might be triggered by the opening of fusion pores between contiguous mitochondria.
Collapse
Affiliation(s)
- Manon Rosselin
- Department of Cell Physiology and Metabolism, University of Geneva, Switzerland
| | - Paula Nunes-Hasler
- Department of Cell Physiology and Metabolism, University of Geneva, Switzerland
| | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, University of Geneva, Switzerland
| |
Collapse
|
26
|
Wang S, Hu M, He H. Quantitative analysis of mitoflash excited by femtosecond laser. JOURNAL OF BIOMEDICAL OPTICS 2018; 23:1-6. [PMID: 29952149 DOI: 10.1117/1.jbo.23.6.065005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/13/2018] [Indexed: 06/08/2023]
Abstract
Mitochondrial oxidative flashes (mitoflashes) are oxidative burst events in mitochondria. It is crosslinked with numerous mitochondrial molecular processes and related with pivotal cell functions such as apoptosis and respiration. In previous research, mitoflashes were found as spontaneous occasional events. It would be observed more frequently if cells were treated with proapoptotic chemicals. We show that multiple mitoflashes can be initiated by a single femtosecond-laser stimulation that was tightly focused on a diffraction-limited spot in the mitochondrial tubular structure. The mitoflash events triggered by different photostimulations are quantified and analyzed. The width and amplitude of mitoflashes are found very sensitive to stimulation parameters including laser power, exposure duration, and total incident laser energy. This study provides a quantitative investigation on the photostimulated mitoflashes. It may thus demonstrate such optical method to be a promising technique in future mitochondrial research.
Collapse
Affiliation(s)
| | | | - Hao He
- Tianjin Univ., China
- Shanghai Jiao Tong Univ., China
| |
Collapse
|
27
|
Chen W, Luo S, Xie P, Hou T, Yu T, Fu X. Overexpressed UCP2 regulates mitochondrial flashes and reverses lipopolysaccharide-induced cardiomyocytes injury. Am J Transl Res 2018; 10:1347-1356. [PMID: 29887950 PMCID: PMC5992536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/06/2018] [Indexed: 06/08/2023]
Abstract
Background: Mitochondrial flashes (mitoflashes) are transient signals from transient bursts of reactive oxygen species (ROS) and changes in pH that occur in certain physiological or pathological conditions. Mitoflashes are closely related to metabolism, cell differentiation, stress response, diseases, and aging. Sepsis can trigger mitochondrial dysfunction in myocardial cells, which leads to ROS overproduction, while uncoupling protein 2 (UCP2) can reduce ROS production. This study aims to observe whether UCP2 overexpression can regulate the frequency of mitoflashes in cardiomyocytes during sepsis and thereby play a protective role. Methods: A cell model for sepsis-induced myocardial damage was established using lipopolysaccharide (LPS). UCP2 overexpression in cardiomyocytes was achieved by adenovirus transfection. Creatinine kinase (CK), lactate dehydrogenase (LDH), tumor necrosis factor (TNF-α), and interleukin (IL-6) activities were detected, and mitochondrial membrane potentials (MMP) were measured. The frequency of mitoflashes in cardiomyocytes was observed. Results: With LPS stimulation, mitoflashes in cardiomyocytes increased significantly, and the MMP was damaged. Additionally, significant increases in CK, LDH, TNF-α, and IL-6 expression levels were observed. UCP2 overexpression can significantly reverse myocardial cell injuries that result from LPS stimulation. Compared with the LPS group, the LPS+UCP2 overexpression group showed a decrease in mitoflash frequency, an improved MMP, and decreases in CK, LDH, TNF-α, and IL-6 expression levels. Conclusion: This study is the first to demonstrate the function of UCP2 overexpression in protecting the myocardium by regulating mitoflash frequency and reversing sepsis-induced myocardial injuries.
Collapse
Affiliation(s)
- Wenbo Chen
- Department of Critical Care Medicine, The First Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Shiyu Luo
- Department of Critical Care Medicine, The First Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Peng Xie
- Department of Critical Care Medicine, The First Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| | - Tingting Hou
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking UniversityBeijing 100871, China
| | - Tian Yu
- Guizhou Key Laboratory of Anesthesia and Organ Protection, Zunyi Medical UniversityZunyi, Guizhou, China
| | - Xiaoyun Fu
- Department of Critical Care Medicine, The First Affiliated Hospital of Zunyi Medical UniversityZunyi, Guizhou, China
| |
Collapse
|
28
|
Cheng AJ, Place N, Westerblad H. Molecular Basis for Exercise-Induced Fatigue: The Importance of Strictly Controlled Cellular Ca 2+ Handling. Cold Spring Harb Perspect Med 2018; 8:cshperspect.a029710. [PMID: 28432118 DOI: 10.1101/cshperspect.a029710] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The contractile function of skeletal muscle declines during intense or prolonged physical exercise, that is, fatigue develops. Skeletal muscle fibers fatigue acutely during highly intense exercise when they have to rely on anaerobic metabolism. Early stages of fatigue involve impaired myofibrillar function, whereas decreased Ca2+ release from the sarcoplasmic reticulum (SR) becomes more important in later stages. SR Ca2+ release can also become reduced with more prolonged, lower intensity exercise, and it is then related to glycogen depletion. Increased reactive oxygen/nitrogen species can cause long-lasting impairments in SR Ca2+ release resulting in a prolonged force depression after exercise. In this article, we discuss molecular and cellular mechanisms of the above fatigue-induced changes, with special focus on multiple mechanisms to decrease SR Ca2+ release to avoid energy depletion and preserve muscle fiber integrity. We also discuss fatigue-related effects of exercise-induced Ca2+ fluxes over the sarcolemma and between the cytoplasm and mitochondria.
Collapse
Affiliation(s)
- Arthur J Cheng
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Nicolas Place
- Institute of Sport Sciences, Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
| | - Håkan Westerblad
- Department of Physiology and Pharmacology, Karolinska Institutet, 171 77 Stockholm, Sweden
| |
Collapse
|
29
|
Wang W, Fernandez-Sanz C, Sheu SS. Regulation of mitochondrial bioenergetics by the non-canonical roles of mitochondrial dynamics proteins in the heart. Biochim Biophys Acta Mol Basis Dis 2017; 1864:1991-2001. [PMID: 28918113 DOI: 10.1016/j.bbadis.2017.09.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 08/25/2017] [Accepted: 09/05/2017] [Indexed: 01/09/2023]
Abstract
Recent advancement in mitochondrial research has significantly extended our knowledge on the role and regulation of mitochondria in health and disease. One important breakthrough is the delineation of how mitochondrial morphological changes, termed mitochondrial dynamics, are coupled to the bioenergetics and signaling functions of mitochondria. In general, it is believed that fusion leads to an increased mitochondrial respiration efficiency and resistance to stress-induced dysfunction while fission does the contrary. This concept seems not applicable to adult cardiomyocytes. The mitochondria in adult cardiomyocytes exhibit fragmented morphology (tilted towards fission) and show less networking and movement as compared to other cell types. However, being the most energy-demanding cells, cardiomyocytes in the adult heart possess vast number of mitochondria, high level of energy flow, and abundant mitochondrial dynamics proteins. This apparent discrepancy could be explained by recently identified new functions of the mitochondrial dynamics proteins. These "non-canonical" roles of mitochondrial dynamics proteins range from controlling inter-organelle communication to regulating cell viability and survival under metabolic stresses. Here, we summarize the newly identified non-canonical roles of mitochondrial dynamics proteins. We focus on how these fission and fusion independent roles of dynamics proteins regulate mitochondrial bioenergetics. We also discuss potential molecular mechanisms, unique intracellular location, and the cardiovascular disease relevance of these non-canonical roles of the dynamics proteins. We propose that future studies are warranted to differentiate the canonical and non-canonical roles of dynamics proteins and to identify new approaches for the treatment of heart diseases. This article is part of a Special issue entitled Cardiac adaptations to obesity, diabetes and insulin resistance, edited by Professors Jan F.C. Glatz, Jason R.B. Dyck and Christine Des Rosiers.
Collapse
Affiliation(s)
- Wang Wang
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA.
| | - Celia Fernandez-Sanz
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
30
|
Antioxidant Treatment Reduces Formation of Structural Cores and Improves Muscle Function in RYR1 Y522S/WT Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6792694. [PMID: 29062463 PMCID: PMC5610828 DOI: 10.1155/2017/6792694] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 06/13/2017] [Indexed: 12/27/2022]
Abstract
Central core disease (CCD) is a congenital myopathy linked to mutations in the ryanodine receptor type 1 (RYR1), the sarcoplasmic reticulum Ca2+ release channel of skeletal muscle. CCD is characterized by formation of amorphous cores within muscle fibers, lacking mitochondrial activity. In skeletal muscle of RYR1Y522S/WT knock-in mice, carrying a human mutation in RYR1 linked to malignant hyperthermia (MH) with cores, oxidative stress is elevated and fibers present severe mitochondrial damage and cores. We treated RYR1Y522S/WT mice with N-acetylcysteine (NAC), an antioxidant provided ad libitum in drinking water for either 2 or 6 months. Our results show that 2 months of NAC treatment starting at 2 months of age, when mitochondrial and fiber damage was still minimal, (i) reduce formation of unstructured and contracture cores, (ii) improve muscle function, and (iii) decrease mitochondrial damage. The beneficial effect of NAC treatment is also evident following 6 months of treatment starting at 4 months of age, when structural damage was at an advanced stage. NAC exerts its protective effect likely by lowering oxidative stress, as supported by the reduction of 3-NT and SOD2 levels. This work suggests that NAC administration is beneficial to prevent mitochondrial damage and formation of cores and improve muscle function in RYR1Y522S/WT mice.
Collapse
|
31
|
Wang X, Zhang X, Wu D, Huang Z, Hou T, Jian C, Yu P, Lu F, Zhang R, Sun T, Li J, Qi W, Wang Y, Gao F, Cheng H. Mitochondrial flashes regulate ATP homeostasis in the heart. eLife 2017; 6. [PMID: 28692422 PMCID: PMC5503511 DOI: 10.7554/elife.23908] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 05/16/2017] [Indexed: 01/01/2023] Open
Abstract
The maintenance of a constant ATP level (‘set-point’) is a vital homeostatic function shared by eukaryotic cells. In particular, mammalian myocardium exquisitely safeguards its ATP set-point despite 10-fold fluctuations in cardiac workload. However, the exact mechanisms underlying this regulation of ATP homeostasis remain elusive. Here we show mitochondrial flashes (mitoflashes), recently discovered dynamic activity of mitochondria, play an essential role for the auto-regulation of ATP set-point in the heart. Specifically, mitoflashes negatively regulate ATP production in isolated respiring mitochondria and, their activity waxes and wanes to counteract the ATP supply-demand imbalance caused by superfluous substrate and altered workload in cardiomyocytes. Moreover, manipulating mitoflash activity is sufficient to inversely shift the otherwise stable ATP set-point. Mechanistically, the Bcl-xL-regulated proton leakage through F1Fo-ATP synthase appears to mediate the coupling between mitoflash production and ATP set-point regulation. These findings indicate mitoflashes appear to constitute a digital auto-regulator for ATP homeostasis in the heart. DOI:http://dx.doi.org/10.7554/eLife.23908.001 A small molecule called ATP is often referred to as the primary “energy currency” of living cells. It is required to power tasks as diverse as the general housekeeping processes that keep all cells alive to the programmed cell death response that dismantles any cells that are no longer needed. It is also crucial that cells maintain a constant level of ATP at all times, even when the supply of and demand for ATP fluctuate. This control is particularly important in the mammalian heart where the rates of ATP production and consumption change ten-fold during intense exercise. Despite intensive research over the past decades, it was still not known how cells keep ATP levels constant. In many cell types, including heart muscle cells, ATP is mainly produced inside compartments called mitochondria. Each heart muscle cell contains between 5,000 and 8,000 mitochondria. Recent experiments have shown that ATP production in mitochondria is interrupted by ten-second bursts called “mitochondrial flashes” (or mitoflashes for short), during which the mitochondria release chemicals called reactive oxygen species. The mitoflashes are tightly linked with energy usage, and Wang, Zhang, Wu et al. have now explored if and how mitoflashes regulate ATP levels in the heart. Experiments on isolated mitochondria from mouse heart muscle cells showed that mitoflashes inhibit the production of ATP. When the intact heart muscle cells were given excess of the building blocks needed to produce ATP – mitoflashes occurred more often. Conversely, when the cells were forced to contract more quickly, which increased demand for ATP, the mitoflashes occurred less often. Importantly, the level of ATP inside the cells actually remained constant in the experiments. Furthermore, inhibiting mitoflashes with antioxidants increased the ATP concentration in heart muscle cells. Lastly, Wang et al. demonstrated that mitoflashes could be triggered under certain conditions. Overall, these experiments uncovered a way in which highly active cells can maintain a constant level of ATP. Future studies are needed to understand exactly how mitoflashes are initiated and how they in turn inhibit ATP production. A better understanding of these processes might uncover molecules that could be targeted by drugs to the control of the rate of ATP production to treat heart failure. DOI:http://dx.doi.org/10.7554/eLife.23908.002
Collapse
Affiliation(s)
- Xianhua Wang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xing Zhang
- Department of Aerospace Medicine, The Fourth Military Medical University, Xi'an, China
| | - Di Wu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Zhanglong Huang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Tingting Hou
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Chongshu Jian
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Peng Yu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Fujian Lu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Rufeng Zhang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Tao Sun
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Jinghang Li
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Wenfeng Qi
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Yanru Wang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Feng Gao
- Department of Aerospace Medicine, The Fourth Military Medical University, Xi'an, China
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| |
Collapse
|
32
|
Fu ZX, Tan X, Fang H, Lau PM, Wang X, Cheng H, Bi GQ. Dendritic mitoflash as a putative signal for stabilizing long-term synaptic plasticity. Nat Commun 2017; 8:31. [PMID: 28652625 PMCID: PMC5484698 DOI: 10.1038/s41467-017-00043-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 04/28/2017] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial flashes (mitoflashes) are recently discovered excitable mitochondrial events in many cell types. Here we investigate their occurrence in the context of structural long-term potentiation (sLTP) at hippocampal synapses. At dendritic spines stimulated by electric pulses, glycine, or targeted glutamate uncaging, induction of sLTP is associated with a phasic occurrence of local, quantized mitochondrial activity in the form of one or a few mitoflashes, over a 30-min window. Low-dose nigericin or photoactivation that elicits mitoflashes stabilizes otherwise short-term spine enlargement into sLTP. Meanwhile, scavengers of reactive oxygen species suppress mitoflashes while blocking sLTP. With targeted photoactivation of mitoflashes, we further show that the stabilization of sLTP is effective within the critical 30-min time-window and a spatial extent of ~2 μm, similar to that of local diffusive reactive oxygen species. These findings indicate a potential signaling role of dendritic mitochondria in synaptic plasticity, and provide new insights into the cellular function of mitoflashes. Mitoflashes are dynamic events in mitochondria, associated with depolarization and release of reactive oxygen species, and have been associated with several cellular functions. The authors now show that in neurons, dendritic mitoflashes are involved in structural postsynaptic changes during LTP.
Collapse
Affiliation(s)
- Zhong-Xiao Fu
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230027, China.,School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.,State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Xiao Tan
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.,State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.,CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, 230027, China
| | - Huaqiang Fang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China
| | - Pak-Ming Lau
- School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China.,CAS Key Laboratory of Brain Function and Disease, University of Science and Technology of China, Hefei, 230027, China
| | - Xianhua Wang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China. .,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China. .,Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, China.
| | - Guo-Qiang Bi
- Hefei National Laboratory for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei, 230027, China. .,School of Life Sciences, University of Science and Technology of China, Hefei, 230027, China. .,CAS Center for Excellence in Brain Science and Intelligence Technology, University of Science and Technology of China, Hefei, 230027, China. .,Innovation Center for Cell Signaling Network, University of Science and Technology of China, Hefei, 230027, China.
| |
Collapse
|
33
|
Jian C, Xu F, Hou T, Sun T, Li J, Cheng H, Wang X. Deficiency of PHB complex impairs respiratory supercomplex formation and activates mitochondrial flashes. J Cell Sci 2017. [PMID: 28630166 DOI: 10.1242/jcs.198523] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Prohibitins (PHBs; prohibitin 1, PHB1 or PHB, and prohibitin 2, PHB2) are evolutionarily conserved and ubiquitously expressed mitochondrial proteins. PHBs form multimeric ring complexes acting as scaffolds in the inner mitochondrial membrane. Mitochondrial flashes (mitoflashes) are newly discovered mitochondrial signaling events that reflect electrical and chemical excitations of the organelle. Here, we investigate the possible roles of PHBs in the regulation of mitoflash signaling. Downregulation of PHBs increases mitoflash frequency by up to 5.4-fold due to elevated basal reactive oxygen species (ROS) production in the mitochondria. Mechanistically, PHB deficiency impairs the formation of mitochondrial respiratory supercomplexes (RSCs) without altering the abundance of individual respiratory complex subunits. These impairments induced by PHB deficiency are effectively rescued by co-expression of PHB1 and PHB2, indicating that the multimeric PHB complex acts as the functional unit. Furthermore, downregulating other RSC assembly factors, including SCAFI (also known as COX7A2L), RCF1a (HIGD1A), RCF1b (HIGD2A), UQCC3 and SLP2 (STOML2), all activate mitoflashes through elevating mitochondrial ROS production. Our findings identify the PHB complex as a new regulator of RSC formation and mitoflash signaling, and delineate a general relationship among RSC formation, basal ROS production and mitoflash biogenesis.
Collapse
Affiliation(s)
- Chongshu Jian
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Fengli Xu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Tingting Hou
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Tao Sun
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Jinghang Li
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xianhua Wang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| |
Collapse
|
34
|
Wang X, Zhang X, Huang Z, Wu D, Liu B, Zhang R, Yin R, Hou T, Jian C, Xu J, Zhao Y, Wang Y, Gao F, Cheng H. Protons Trigger Mitochondrial Flashes. Biophys J 2017; 111:386-394. [PMID: 27463140 DOI: 10.1016/j.bpj.2016.05.052] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 05/06/2016] [Accepted: 05/31/2016] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence indicates that mitochondrial flashes (mitoflashes) are highly conserved elemental mitochondrial signaling events. However, which signal controls their ignition and how they are integrated with other mitochondrial signals and functions remain elusive. In this study, we aimed to further delineate the signal components of the mitoflash and determine the mitoflash trigger mechanism. Using multiple biosensors and chemical probes as well as label-free autofluorescence, we found that the mitoflash reflects chemical and electrical excitation at the single-organelle level, comprising bursting superoxide production, oxidative redox shift, and matrix alkalinization as well as transient membrane depolarization. Both electroneutral H(+)/K(+) or H(+)/Na(+) antiport and matrix proton uncaging elicited immediate and robust mitoflash responses over a broad dynamic range in cardiomyocytes and HeLa cells. However, charge-uncompensated proton transport, which depolarizes mitochondria, caused the opposite effect, and steady matrix acidification mildly inhibited mitoflashes. Based on a numerical simulation, we estimated a mean proton lifetime of 1.42 ns and diffusion distance of 2.06 nm in the matrix. We conclude that nanodomain protons act as a novel, to our knowledge, trigger of mitoflashes in energized mitochondria. This finding suggests that mitoflash genesis is functionally and mechanistically integrated with mitochondrial energy metabolism.
Collapse
Affiliation(s)
- Xianhua Wang
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China.
| | - Xing Zhang
- Department of Aerospace Medicine, The Fourth Military Medical University, Xi'an, China
| | - Zhanglong Huang
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Di Wu
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Beibei Liu
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Rufeng Zhang
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Rongkang Yin
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Tingting Hou
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Chongshu Jian
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Jiejia Xu
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Yan Zhao
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Yanru Wang
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Feng Gao
- Department of Aerospace Medicine, The Fourth Military Medical University, Xi'an, China
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China.
| |
Collapse
|
35
|
Michelucci A, Paolini C, Boncompagni S, Canato M, Reggiani C, Protasi F. Strenuous exercise triggers a life-threatening response in mice susceptible to malignant hyperthermia. FASEB J 2017; 31:3649-3662. [PMID: 28465322 DOI: 10.1096/fj.201601292r] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 04/17/2017] [Indexed: 12/26/2022]
Abstract
In humans, hyperthermic episodes can be triggered by halogenated anesthetics [malignant hyperthermia (MH) susceptibility] and by high temperature [environmental heat stroke (HS)]. Correlation between MH susceptibility and HS is supported by extensive work in mouse models that carry a mutation in ryanodine receptor type-1 (RYR1Y522S/WT) and calsequestrin-1 knockout (CASQ1-null), 2 proteins that control Ca2+ release in skeletal muscle. As overheating episodes in humans have also been described during exertion, here we subjected RYR1Y522S/WT and CASQ1-null mice to an exertional-stress protocol (incremental running on a treadmill at 34°C and 40% humidity). The mortality rate was 80 and 78.6% in RYR1Y522S/WT and CASQ1-null mice, respectively, vs. 0% in wild-type mice. Lethal crises were characterized by hyperthermia and rhabdomyolysis, classic features of MH episodes. Of importance, pretreatment with azumolene, an analog of the drug used in humans to treat MH crises, reduced mortality to 0 and 12.5% in RYR1Y522S/WT and CASQ1-null mice, respectively, thanks to a striking reduction of hyperthermia and rhabdomyolysis. At the molecular level, azumolene strongly prevented Ca2+-dependent activation of calpains and NF-κB by lowering myoplasmic Ca2+ concentration and nitro-oxidative stress, parameters that were elevated in RYR1Y522S/WT and CASQ1-null mice. These results suggest that common molecular mechanisms underlie MH crises and exertional HS in mice.-Michelucci, A., Paolini, C., Boncompagni, S., Canato, M., Reggiani, C., Protasi, F. Strenuous exercise triggers a life-threatening response in mice susceptible to malignant hyperthermia.
Collapse
Affiliation(s)
- Antonio Michelucci
- Center for Research on Ageing and Translational Medicine (CeSI-MeT), Department of Neuroscience, Imaging, and Clinical Sciences (DNICS), Università degli Studi G. d'Annunzio, Chieti, Italy
| | - Cecilia Paolini
- Center for Research on Ageing and Translational Medicine (CeSI-MeT), Department of Neuroscience, Imaging, and Clinical Sciences (DNICS), Università degli Studi G. d'Annunzio, Chieti, Italy
| | - Simona Boncompagni
- Center for Research on Ageing and Translational Medicine (CeSI-MeT), Department of Neuroscience, Imaging, and Clinical Sciences (DNICS), Università degli Studi G. d'Annunzio, Chieti, Italy
| | - Marta Canato
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Carlo Reggiani
- Department of Biomedical Sciences, University of Padova, Padua, Italy
| | - Feliciano Protasi
- Center for Research on Ageing and Translational Medicine (CeSI-MeT), Department of Neuroscience, Imaging, and Clinical Sciences (DNICS), Università degli Studi G. d'Annunzio, Chieti, Italy; .,Department of Medicine and Aging Science, University G. d' Annunzio of Chieti, Chieti, Italy
| |
Collapse
|
36
|
Karam C, Yi J, Xiao Y, Dhakal K, Zhang L, Li X, Manno C, Xu J, Li K, Cheng H, Ma J, Zhou J. Absence of physiological Ca 2+ transients is an initial trigger for mitochondrial dysfunction in skeletal muscle following denervation. Skelet Muscle 2017; 7:6. [PMID: 28395670 PMCID: PMC5387329 DOI: 10.1186/s13395-017-0123-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 03/08/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Motor neurons control muscle contraction by initiating action potentials in muscle. Denervation of muscle from motor neurons leads to muscle atrophy, which is linked to mitochondrial dysfunction. It is known that denervation promotes mitochondrial reactive oxygen species (ROS) production in muscle, whereas the initial cause of mitochondrial ROS production in denervated muscle remains elusive. Since denervation isolates muscle from motor neurons and deprives it from any electric stimulation, no action potentials are initiated, and therefore, no physiological Ca2+ transients are generated inside denervated muscle fibers. We tested whether loss of physiological Ca2+ transients is an initial cause leading to mitochondrial dysfunction in denervated skeletal muscle. METHODS A transgenic mouse model expressing a mitochondrial targeted biosensor (mt-cpYFP) allowed a real-time measurement of the ROS-related mitochondrial metabolic function following denervation, termed "mitoflash." Using live cell imaging, electrophysiological, pharmacological, and biochemical studies, we examined a potential molecular mechanism that initiates ROS-related mitochondrial dysfunction following denervation. RESULTS We found that muscle fibers showed a fourfold increase in mitoflash activity 24 h after denervation. The denervation-induced mitoflash activity was likely associated with an increased activity of mitochondrial permeability transition pore (mPTP), as the mitoflash activity was attenuated by application of cyclosporine A. Electrical stimulation rapidly reduced mitoflash activity in both sham and denervated muscle fibers. We further demonstrated that the Ca2+ level inside mitochondria follows the time course of the cytosolic Ca2+ transient and that inhibition of mitochondrial Ca2+ uptake by Ru360 blocks the effect of electric stimulation on mitoflash activity. CONCLUSIONS The loss of cytosolic Ca2+ transients due to denervation results in the downstream absence of mitochondrial Ca2+ uptake. Our studies suggest that this could be an initial trigger for enhanced mPTP-related mitochondrial ROS generation in skeletal muscle.
Collapse
Affiliation(s)
- Chehade Karam
- Rush University School of Medicine, Chicago, IL, USA
| | - Jianxun Yi
- Rush University School of Medicine, Chicago, IL, USA.,Kansas City University of Medicine and Bioscience, 1750 Independence Ave, Kansas City, MO, 64106, USA
| | - Yajuan Xiao
- Rush University School of Medicine, Chicago, IL, USA.,Kansas City University of Medicine and Bioscience, 1750 Independence Ave, Kansas City, MO, 64106, USA
| | - Kamal Dhakal
- Kansas City University of Medicine and Bioscience, 1750 Independence Ave, Kansas City, MO, 64106, USA
| | - Lin Zhang
- Rush University School of Medicine, Chicago, IL, USA.,Kansas City University of Medicine and Bioscience, 1750 Independence Ave, Kansas City, MO, 64106, USA
| | - Xuejun Li
- Kansas City University of Medicine and Bioscience, 1750 Independence Ave, Kansas City, MO, 64106, USA
| | - Carlo Manno
- Rush University School of Medicine, Chicago, IL, USA
| | - Jiejia Xu
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - Kaitao Li
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - Heping Cheng
- Institute of Molecular Medicine, Peking University, Beijing, China
| | - Jianjie Ma
- Wexner Medical Center, The Ohio State University, 460 West 12th Avenue, Columbus, OH, USA.
| | - Jingsong Zhou
- Rush University School of Medicine, Chicago, IL, USA. .,Kansas City University of Medicine and Bioscience, 1750 Independence Ave, Kansas City, MO, 64106, USA.
| |
Collapse
|
37
|
Rosselin M, Santo-Domingo J, Bermont F, Giacomello M, Demaurex N. L-OPA1 regulates mitoflash biogenesis independently from membrane fusion. EMBO Rep 2017; 18:451-463. [PMID: 28174208 PMCID: PMC5331265 DOI: 10.15252/embr.201642931] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2016] [Revised: 12/19/2016] [Accepted: 12/21/2016] [Indexed: 11/09/2022] Open
Abstract
Mitochondrial flashes mediated by optic atrophy 1 (OPA1) fusion protein are bioenergetic responses to stochastic drops in mitochondrial membrane potential (Δψm) whose origin is unclear. Using structurally distinct genetically encoded pH‐sensitive probes, we confirm that flashes are matrix alkalinization transients, thereby establishing the pH nature of these events, which we renamed “mitopHlashes”. Probes located in cristae or intermembrane space as verified by electron microscopy do not report pH changes during Δψm drops or respiratory chain inhibition. Opa1 ablation does not alter Δψm fluctuations but drastically decreases the efficiency of mitopHlash/Δψm coupling, which is restored by re‐expressing fusion‐deficient OPA1K301A and preserved in cells lacking the outer‐membrane fusion proteins MFN1/2 or the OPA1 proteases OMA1 and YME1L, indicating that mitochondrial membrane fusion and OPA1 proteolytic processing are dispensable. pH/Δψm uncoupling occurs early during staurosporine‐induced apoptosis and is mitigated by OPA1 overexpression, suggesting that OPA1 maintains mitopHlash competence during stress conditions. We propose that OPA1 stabilizes respiratory chain supercomplexes in a conformation that enables respiring mitochondria to compensate a drop in Δψm by an explosive matrix pH flash.
Collapse
Affiliation(s)
- Manon Rosselin
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | - Jaime Santo-Domingo
- Nestlé Institute of Health Sciences SA, EPFL Innovation Park, Lausanne, Switzerland
| | - Flavien Bermont
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| | | | - Nicolas Demaurex
- Department of Cell Physiology and Metabolism, University of Geneva, Geneva, Switzerland
| |
Collapse
|
38
|
Feng G, Liu B, Hou T, Wang X, Cheng H. Mitochondrial Flashes: Elemental Signaling Events in Eukaryotic Cells. Handb Exp Pharmacol 2017; 240:403-422. [PMID: 28233181 DOI: 10.1007/164_2016_129] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Mitochondrial flashes (mitoflashes) are recently discovered mitochondrial activity which reflects chemical and electrical excitation of the organelle. Emerging evidence indicates that mitoflashes represent highly regulated, elementary signaling events that play important roles in physiological and pathophysiological processes in eukaryotes. Furthermore, they are regulated by mitochondrial ROS, Ca2+, and protons, and are intertwined with mitochondrial metabolic processes. As such, targeting mitoflash activity may provide a novel means for the control of mitochondrial metabolism and signaling in health and disease. In this brief review, we summarize salient features and mechanisms of biogenesis of mitoflashes, and synthesize data on mitoflash biology in the context of metabolism, cell differentiation, stress response, disease, and ageing.
Collapse
Affiliation(s)
- Gaomin Feng
- Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Beibei Liu
- Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Tingting Hou
- Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Xianhua Wang
- Institute of Molecular Medicine, Peking University, Beijing, 100871, China
| | - Heping Cheng
- Institute of Molecular Medicine, Peking University, Beijing, 100871, China.
| |
Collapse
|
39
|
McMurray F, Patten DA, Harper ME. Reactive Oxygen Species and Oxidative Stress in Obesity-Recent Findings and Empirical Approaches. Obesity (Silver Spring) 2016; 24:2301-2310. [PMID: 27804267 DOI: 10.1002/oby.21654] [Citation(s) in RCA: 176] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/26/2016] [Accepted: 08/01/2016] [Indexed: 12/12/2022]
Abstract
OBJECTIVE High levels of reactive oxygen species (ROS) are intricately linked to obesity and associated pathologies, notably insulin resistance and type 2 diabetes. However, ROS are also thought to be important in intracellular signaling, which may paradoxically be required for insulin sensitivity. Many theories have been developed to explain this apparent paradox, which have broadened our understanding of these important small molecules. While many sites for intracellular ROS production have been described, mitochondrial generated ROS remain a major contributor in most cell types. Mitochondrial ROS generation is controlled by a number of factors described in this review. Moreover, these studies have established both a demand for novel sensitive approaches to measure ROS, as well as a need to standardize and review their suitability for different applications. METHODS To properly assess levels of ROS and mitochondrial ROS in the development of obesity and its complications, a growing number of tools have been developed. This paper reviews many of the common methods for the investigation of ROS in mitochondria, cell, animal, and human models. RESULTS Available approaches can be generally divided into those that measure ROS-induced damage (e.g., DNA, lipid, and protein damage); those that measure antioxidant levels and redox ratios; and those that use novel biosensors and probes for a more direct measure of different forms of ROS (e.g., 2',7'-di-chlorofluorescein (DCF), dihydroethidium (DHE) and its mitochondrial targeted form (MitoSOX), Amplex Red, roGFP, HyPer, mt-cpYFP, ratiometric H2 O2 probes, and their derivatives). Moreover, this review provides caveats and strengths for the use of these techniques in different models. CONCLUSIONS Advances in these techniques will undoubtedly advance the understanding of ROS in obesity and may help resolve unanswered questions in the field.
Collapse
Affiliation(s)
- Fiona McMurray
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - David A Patten
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Mary-Ellen Harper
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| |
Collapse
|
40
|
|
41
|
Pietrangelo L, D'Incecco A, Ainbinder A, Michelucci A, Kern H, Dirksen RT, Boncompagni S, Protasi F. Age-dependent uncoupling of mitochondria from Ca2⁺ release units in skeletal muscle. Oncotarget 2016; 6:35358-71. [PMID: 26485763 PMCID: PMC4742110 DOI: 10.18632/oncotarget.6139] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Accepted: 09/22/2015] [Indexed: 12/03/2022] Open
Abstract
Calcium release units (CRUs) and mitochondria control myoplasmic [Ca2+] levels and ATP production in muscle, respectively. We recently reported that these two organelles are structurally connected by tethers, which promote proximity and proper Ca2+ signaling. Here we show that disposition, ultrastructure, and density of CRUs and mitochondria and their reciprocal association are compromised in muscle from aged mice. Specifically, the density of CRUs and mitochondria is decreased in muscle fibers from aged (>24 months) vs. adult (3-12 months), with an increased percentage of mitochondria being damaged and misplaced from their normal triadic position. A significant reduction in tether (13.8±0.4 vs. 5.5±0.3 tethers/100μm2) and CRU-mitochondrial pair density (37.4±0.8 vs. 27.0±0.7 pairs/100μm2) was also observed in aged mice. In addition, myoplasmic Ca2+ transient (1.68±0.08 vs 1.37±0.03) and mitochondrial Ca2+ uptake (9.6±0.050 vs 6.58±0.54) during repetitive high frequency tetanic stimulation were significantly decreased. Finally oxidative stress, assessed from levels of 3-nitrotyrosine (3-NT), Cu/Zn superoxide-dismutase (SOD1) and Mn superoxide dismutase (SOD2) expression, were significantly increased in aged mice. The reduced association between CRUs and mitochondria with aging may contribute to impaired cross-talk between the two organelles, possibly resulting in reduced efficiency in activity-dependent ATP production and, thus, to age-dependent decline of skeletal muscle performance.
Collapse
Affiliation(s)
- Laura Pietrangelo
- CeSI - Center for Research on Aging & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio, Chieti, Italy
| | - Alessandra D'Incecco
- CeSI - Center for Research on Aging & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio, Chieti, Italy
| | - Alina Ainbinder
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Antonio Michelucci
- CeSI - Center for Research on Aging & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio, Chieti, Italy
| | - Helmut Kern
- Ludwig Boltzmann Institute of Electrical Stimulation and Physical Rehabilitation & Institute of Physical Medicine and Rehabilitation, Wilhelminenspital, Vienna, Austria
| | - Robert T Dirksen
- Department of Pharmacology and Physiology, University of Rochester School of Medicine and Dentistry, Rochester, NY, USA
| | - Simona Boncompagni
- CeSI - Center for Research on Aging & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio, Chieti, Italy
| | - Feliciano Protasi
- CeSI - Center for Research on Aging & DNICS, Department of Neuroscience, Imaging and Clinical Sciences, University G. d'Annunzio, Chieti, Italy
| |
Collapse
|
42
|
Wang W, Zhang H, Cheng H. Mitochondrial flashes: From indicator characterization to in vivo imaging. Methods 2016; 109:12-20. [PMID: 27288722 PMCID: PMC5075495 DOI: 10.1016/j.ymeth.2016.06.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 06/03/2016] [Accepted: 06/07/2016] [Indexed: 12/15/2022] Open
Abstract
Mitochondrion is an organelle critically responsible for energy production and intracellular signaling in eukaryotic cells and its dysfunction often accompanies and contributes to human disease. Superoxide is the primary reactive oxygen species (ROS) produced in mitochondria. In vivo detection of superoxide has been a challenge in biomedical research. Here we describe the methods used to characterize a circularly permuted yellow fluorescent protein (cpYFP) as a biosensor for mitochondrial superoxide and pH dynamics. In vitro characterization reveals the high selectivity of cpYFP to superoxide over other ROS species and its dual sensitivity to pH. Confocal and two-photon imaging in conjunction with transgenic expression of the biosensor cpYFP targeted to the mitochondrial matrix detects mitochondrial flash events in living cells, perfused intact hearts, and live animals. The mitochondrial flashes are discrete and stochastic single mitochondrial events triggered by transient mitochondrial permeability transition (tMPT) and composed of a bursting superoxide signal and a transient alkalization signal. The real-time monitoring of single mitochondrial flashes provides a unique tool to study the integrated dynamism of mitochondrial respiration, ROS production, pH regulation and tMPT kinetics under diverse physiological and pathophysiological conditions.
Collapse
Affiliation(s)
- Wang Wang
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA.
| | - Huiliang Zhang
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA 98109, USA
| | - Heping Cheng
- Institute of Molecular Medicine, Peking University, Beijing 100871, China
| |
Collapse
|
43
|
Wang W, Gong G, Wang X, Wei-LaPierre L, Cheng H, Dirksen R, Sheu SS. Mitochondrial Flash: Integrative Reactive Oxygen Species and pH Signals in Cell and Organelle Biology. Antioxid Redox Signal 2016; 25:534-549. [PMID: 27245241 PMCID: PMC5035371 DOI: 10.1089/ars.2016.6739] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 05/27/2016] [Indexed: 01/09/2023]
Abstract
SIGNIFICANCE Recent breakthroughs in mitochondrial research have advanced, reshaped, and revolutionized our view of the role of mitochondria in health and disease. These discoveries include the development of novel tools to probe mitochondrial biology, the molecular identification of mitochondrial functional proteins, and the emergence of new concepts and mechanisms in mitochondrial function regulation. The discovery of "mitochondrial flash" activity has provided unique insights not only into real-time visualization of individual mitochondrial redox and pH dynamics in live cells but has also advanced understanding of the excitability, autonomy, and integration of mitochondrial function in vivo. RECENT ADVANCES The mitochondrial flash is a transient and stochastic event confined within an individual mitochondrion and is observed in a wide range of organisms from plants to Caenorhabditis elegans to mammals. As flash events involve multiple transient concurrent changes within the mitochondrion (e.g., superoxide, pH, and membrane potential), a number of different mitochondrial targeted fluorescent indicators can detect flash activity. Accumulating evidence indicates that flash events reflect integrated snapshots of an intermittent mitochondrial process arising from mitochondrial respiration chain activity associated with the transient opening of the mitochondrial permeability transition pore. CRITICAL ISSUES We review the history of flash discovery, summarize current understanding of flash biology, highlight controversies regarding the relative roles of superoxide and pH signals during a flash event, and bring forth the integration of both signals in flash genesis. FUTURE DIRECTIONS Investigations using flash as a biomarker and establishing its role in cell signaling pathway will move the field forward. Antioxid. Redox Signal. 25, 534-549.
Collapse
Affiliation(s)
- Wang Wang
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, Washington
| | - Guohua Gong
- Department of Anesthesiology and Pain Medicine, Mitochondria and Metabolism Center, University of Washington, Seattle, Washington
| | - Xianhua Wang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Lan Wei-LaPierre
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Robert Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, New York
| | - Shey-Shing Sheu
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| |
Collapse
|
44
|
Li W, Sun T, Liu B, Wu D, Qi W, Wang X, Ma Q, Cheng H. Regulation of Mitoflash Biogenesis and Signaling by Mitochondrial Dynamics. Sci Rep 2016; 6:32933. [PMID: 27623243 PMCID: PMC5020656 DOI: 10.1038/srep32933] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 08/17/2016] [Indexed: 12/30/2022] Open
Abstract
Mitochondria are highly dynamic organelles undergoing constant network reorganization and exhibiting stochastic signaling events in the form of mitochondrial flashes (mitoflashes). Here we investigate whether and how mitochondrial network dynamics regulate mitoflash biogenesis and signaling. We found that mitoflash frequency was largely invariant when network fragmentized or redistributed in the absence of mitofusin (Mfn) 1, Mfn2, or Kif5b. However, Opa1 deficiency decreased spontaneous mitoflash frequency due to superimposing changes in respiratory function, whereas mitoflash response to non-metabolic stimulation was unchanged despite network fragmentation. In Drp1- or Mff-deficient cells whose mitochondria hyperfused into a single whole-cell reticulum, the frequency of mitoflashes of regular amplitude and duration was again unaltered, although brief and low-amplitude “miniflashes” emerged because of improved detection ability. As the network reorganized, however, the signal mass of mitoflash signaling was dynamically regulated in accordance with the degree of network connectivity. These findings demonstrate a novel functional role of mitochondrial network dynamics and uncover a magnitude- rather than frequency-modulatory mechanism in the regulation of mitoflash signaling. In addition, our data support a stochastic trigger model for the ignition of mitoflashes.
Collapse
Affiliation(s)
- Wenwen Li
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Tao Sun
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Beibei Liu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Di Wu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Wenfeng Qi
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Xianhua Wang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Qi Ma
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Institute of Molecular Medicine, Peking University, Beijing, China
| |
Collapse
|
45
|
Cheng AJ, Yamada T, Rassier DE, Andersson DC, Westerblad H, Lanner JT. Reactive oxygen/nitrogen species and contractile function in skeletal muscle during fatigue and recovery. J Physiol 2016; 594:5149-60. [PMID: 26857536 DOI: 10.1113/jp270650] [Citation(s) in RCA: 85] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 12/23/2015] [Indexed: 01/17/2023] Open
Abstract
The production of reactive oxygen/nitrogen species (ROS/RNS) is generally considered to increase during physical exercise. Nevertheless, direct measurements of ROS/RNS often show modest increases in ROS/RNS in muscle fibres even during intensive fatiguing stimulation, and the major source(s) of ROS/RNS during exercise is still being debated. In rested muscle fibres, mild and acute exposure to exogenous ROS/RNS generally increases myofibrillar submaximal force, whereas stronger or prolonged exposure has the opposite effect. Endogenous production of ROS/RNS seems to preferentially decrease submaximal force and positive effects of antioxidants are mainly observed during fatigue induced by submaximal contractions. Fatigued muscle fibres frequently enter a prolonged state of reduced submaximal force, which is caused by a ROS/RNS-dependent decrease in sarcoplasmic reticulum Ca(2+) release and/or myofibrillar Ca(2+) sensitivity. Increased ROS/RNS production during exercise can also be beneficial and recent human and animal studies show that antioxidant supplementation can hamper the beneficial effects of endurance training. In conclusion, increased ROS/RNS production have both beneficial and detrimental effects on skeletal muscle function and the outcome depends on a combination of factors: the type of ROS/RNS; the magnitude, duration and location of ROS/RNS production; and the defence systems, including both endogenous and exogenous antioxidants.
Collapse
Affiliation(s)
| | | | - Dilson E Rassier
- McGill University, 475 Pine Avenue West, Montreal, QC, Canada, H2W1S4
| | | | | | | |
Collapse
|
46
|
Hou T, Jian C, Xu J, Huang AY, Xi J, Hu K, Wei L, Cheng H, Wang X. Identification of EFHD1 as a novel Ca(2+) sensor for mitoflash activation. Cell Calcium 2016; 59:262-70. [PMID: 26975899 DOI: 10.1016/j.ceca.2016.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Revised: 02/23/2016] [Accepted: 03/02/2016] [Indexed: 01/16/2023]
Abstract
Mitochondrial flashes (mitoflashes) represent stochastic and discrete mitochondrial events that each comprises a burst of superoxide production accompanied by transient depolarization and matrix alkalinization in a respiratory mitochondrion. While mitochondrial Ca(2+) is shown to be an important regulator of mitoflash activity, little is known about its specific mechanism of action. Here we sought to determine possible molecular players that mediate the Ca(2+) regulation of mitoflashes by screening mitochondrial proteins containing the Ca(2+)-binding motifs. In silico analysis and targeted siRNA screening identified four mitoflash activators (MICU1, EFHD1, SLC25A23, SLC25A25) and one mitoflash inhibitor (LETM1) in terms of their ability to modulate mitoflash response to hyperosmotic stress. In particular, overexpression or down-regulation of EFHD1 enhanced or depressed mitoflash activation, respectively, under various conditions of mitochondrial Ca(2+) elevations. Yet, it did not alter mitochondrial Ca(2+) handling, mitochondrial respiration, or ROS-induced mitoflash production. Further, disruption of the two EF-hand motifs of EFHD1 abolished its potentiating effect on the mitoflash responses. These results indicate that EFHD1 functions as a novel mitochondrial Ca(2+) sensor underlying Ca(2+)-dependent activation of mitoflashes.
Collapse
Affiliation(s)
- Tingting Hou
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Chongshu Jian
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Jiejia Xu
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - August Yue Huang
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Jianzhong Xi
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China; Department of Biomedical Engineering, College of Engineering, Peking University, Beijing, China
| | - Keping Hu
- Research Center for Pharmacology and Toxicology, Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liping Wei
- Center for Bioinformatics, State Key Laboratory of Protein and Plant Gene Research, School of Life Sciences, Peking University, Beijing, China
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China
| | - Xianhua Wang
- State Key Laboratory of Membrane Biology, Beijing Key Laboratory of Cardiometabolic Molecular Medicine, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, China.
| |
Collapse
|
47
|
Cunha M, Testa E, Komova OV, Nasonova EA, Mel'nikova LA, Shmakova NL, Beuve M. Modeling cell response to low doses of photon irradiation: Part 2--application to radiation-induced chromosomal aberrations in human carcinoma cells. RADIATION AND ENVIRONMENTAL BIOPHYSICS 2016; 55:31-40. [PMID: 26708100 DOI: 10.1007/s00411-015-0622-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 10/11/2015] [Indexed: 06/05/2023]
Abstract
The biological phenomena observed at low doses of ionizing radiation (adaptive response, bystander effects, genomic instability, etc.) are still not well understood. While at high irradiation doses, cellular death may be directly linked to DNA damage, at low doses, other cellular structures may be involved in what are known as non-(DNA)-targeted effects. Mitochondria, in particular, may play a crucial role through their participation in a signaling network involving oxygen/nitrogen radical species. According to the size of the implicated organelles, the fluctuations in the energy deposited into these target structures may impact considerably the response of cells to low doses of ionizing irradiation. Based on a recent simulation of these fluctuations, a theoretical framework was established to have further insight into cell responses to low doses of photon irradiation, namely the triggering of radioresistance mechanisms by energy deposition into specific targets. Three versions of a model are considered depending on the target size and on the number of targets that need to be activated by energy deposition to trigger radioresistance mechanisms. These model versions are applied to the fraction of radiation-induced chromosomal aberrations measured at low doses in human carcinoma cells (CAL51). For this cell line, it was found in the present study that the mechanisms of radioresistance could not be triggered by the activation of a single small target (nanometric size, 100 nm), but could instead be triggered by the activation of a large target (micrometric, 10 μm) or by the activation of a great number of small targets. The mitochondria network, viewed either as a large target or as a set of small units, might be concerned by these low-dose effects.
Collapse
Affiliation(s)
- Micaela Cunha
- Université de Lyon, 69622, Lyon, France
- Université de Lyon 1, Villeurbanne, France
- CNRS/IN2P3, Institut de Physique Nucléaire de Lyon, Villeurbanne, France
| | - Etienne Testa
- Université de Lyon, 69622, Lyon, France
- Université de Lyon 1, Villeurbanne, France
- CNRS/IN2P3, Institut de Physique Nucléaire de Lyon, Villeurbanne, France
| | - Olga V Komova
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Dubna, Moscow Region, Russia
| | - Elena A Nasonova
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Dubna, Moscow Region, Russia
| | - Larisa A Mel'nikova
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Dubna, Moscow Region, Russia
| | - Nina L Shmakova
- Laboratory of Radiation Biology, Joint Institute for Nuclear Research, Dubna, Moscow Region, Russia
| | - Michaël Beuve
- Université de Lyon, 69622, Lyon, France.
- Université de Lyon 1, Villeurbanne, France.
- CNRS/IN2P3, Institut de Physique Nucléaire de Lyon, Villeurbanne, France.
| |
Collapse
|
48
|
Shang W, Gao H, Lu F, Ma Q, Fang H, Sun T, Xu J, Ding Y, Lin Y, Wang Y, Wang X, Cheng H, Zheng M. Cyclophilin D regulates mitochondrial flashes and metabolism in cardiac myocytes. J Mol Cell Cardiol 2016; 91:63-71. [PMID: 26746144 DOI: 10.1016/j.yjmcc.2015.10.036] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 09/01/2015] [Accepted: 10/06/2015] [Indexed: 01/24/2023]
Abstract
Cyclophilin D (CyP-D) is the mitochondrial-specific member of the evolutionally conserved cyclophilin family, and plays an important role in the regulation of mitochondrial permeability transition (MPT) under stress. Recently we have demonstrated that respiratory mitochondria undergo mitochondrial flash ("mitoflash") activity which is coupled with transient MPT under physiological conditions. However, whether and how CyP-D regulates mitoflashes remain incompletely understood. By using both loss- and gain-of-function approaches in isolated cardiomyocytes, beating hearts, and skeletal muscles in living mice, we revisited the role of CyP-D in the regulation of mitoflashes. Overexpression of CyP-D increased, and knockout of it halved, cardiac mitoflash frequency, while mitoflash amplitude and kinetics remained unaffected. However, CyP-D ablation did not alter mitoflash frequency, with mitoflash amplitude increased, in gastrocnemius muscles. This disparity was accompanied by 4-fold higher CyP-D expression in mouse cardiac than skeletal muscles. The mitochondrial maximal respiration rate and reserved capacity were reduced in CyP-D-null cardiomyocytes. These data indicate that CyP-D is a significant regulator of mitoflash ignition and mitochondrial metabolism in heart. In addition, tissue-specific CyP-D expression may partly explain the differential regulation of mitoflashes in the two types of striated muscles.
Collapse
MESH Headings
- Animals
- Bacterial Proteins/genetics
- Bacterial Proteins/metabolism
- Peptidyl-Prolyl Isomerase F
- Cyclophilins/genetics
- Cyclophilins/metabolism
- Female
- Gene Expression Regulation
- Genes, Reporter
- Luminescent Proteins/genetics
- Luminescent Proteins/metabolism
- Male
- Membrane Potential, Mitochondrial
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mitochondria/metabolism
- Mitochondria/ultrastructure
- Mitochondrial Membrane Transport Proteins/genetics
- Mitochondrial Membrane Transport Proteins/metabolism
- Mitochondrial Permeability Transition Pore
- Muscle, Skeletal/metabolism
- Muscle, Skeletal/ultrastructure
- Muscle, Striated/metabolism
- Muscle, Striated/ultrastructure
- Myocardium/metabolism
- Myocardium/ultrastructure
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/ultrastructure
- Organ Culture Techniques
- Organ Specificity
- Primary Cell Culture
- Reactive Oxygen Species/metabolism
- Signal Transduction
Collapse
Affiliation(s)
- Wei Shang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Han Gao
- Department of Physiology and Pathophysiology, Health Science Center, Peking University, Beijing 100191, China
| | - Fujian Lu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Qi Ma
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Huaqiang Fang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Tao Sun
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Jiejia Xu
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yi Ding
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yuan Lin
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Yanru Wang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Xianhua Wang
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Heping Cheng
- State Key Laboratory of Membrane Biology, Institute of Molecular Medicine, Peking-Tsinghua Center for Life Sciences, Peking University, Beijing 100871, China
| | - Ming Zheng
- Department of Physiology and Pathophysiology, Health Science Center, Peking University, Beijing 100191, China.
| |
Collapse
|
49
|
Gong G, Liu X, Zhang H, Sheu SS, Wang W. Mitochondrial flash as a novel biomarker of mitochondrial respiration in the heart. Am J Physiol Heart Circ Physiol 2015; 309:H1166-H1177. [PMID: 26276820 PMCID: PMC4666927 DOI: 10.1152/ajpheart.00462.2015] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 08/13/2015] [Indexed: 11/22/2022]
Abstract
Mitochondrial respiration through electron transport chain (ETC) activity generates ATP and reactive oxygen species in eukaryotic cells. The modulation of mitochondrial respiration in vivo or under physiological conditions remains elusive largely due to the lack of appropriate approach to monitor ETC activity in a real-time manner. Here, we show that ETC-coupled mitochondrial flash is a novel biomarker for monitoring mitochondrial respiration under pathophysiological conditions in cultured adult cardiac myocyte and perfused beating heart. Through real-time confocal imaging, we follow the frequency of a transient bursting fluorescent signal, named mitochondrial flash, from individual mitochondria within intact cells expressing a mitochondrial matrix-targeted probe, mt-cpYFP (mitochondrial-circularly permuted yellow fluorescent protein). This mt-cpYFP recorded mitochondrial flash has been shown to be composed of a major superoxide signal with a minor alkalization signal within the mitochondrial matrix. Through manipulating physiological substrates for mitochondrial respiration, we find a close coupling between flash frequency and the ETC electron flow, as measured by oxygen consumption rate in cardiac myocyte. Stimulating electron flow under physiological conditions increases flash frequency. On the other hand, partially block or slowdown electron flow by inhibiting the F0F1 ATPase, which represents a pathological condition, transiently increases then decreases flash frequency. Limiting electron entrance at complex I by knocking out Ndufs4, an assembling subunit of complex I, suppresses mitochondrial flash activity. These results suggest that mitochondrial electron flow can be monitored by real-time imaging of mitochondrial flash. The mitochondrial flash frequency could be used as a novel biomarker for mitochondrial respiration under physiological and pathological conditions.
Collapse
Affiliation(s)
- Guohua Gong
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington; and
| | - Xiaoyun Liu
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington; and
| | - Huiliang Zhang
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington; and
| | - Shey-Shing Sheu
- Center for Translational Medicine, Department of Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Philadelphia
| | - Wang Wang
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, Washington; and
| |
Collapse
|
50
|
Michelucci A, Paolini C, Canato M, Wei-Lapierre L, Pietrangelo L, De Marco A, Reggiani C, Dirksen RT, Protasi F. Antioxidants protect calsequestrin-1 knockout mice from halothane- and heat-induced sudden death. Anesthesiology 2015; 123:603-17. [PMID: 26132720 PMCID: PMC4543432 DOI: 10.1097/aln.0000000000000748] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Mice lacking calsequestrin-1 (CASQ1-null), a Ca-binding protein that modulates the activity of Ca release in the skeletal muscle, exhibit lethal hypermetabolic episodes that resemble malignant hyperthermia in humans when exposed to halothane or heat stress. METHODS Because oxidative species may play a critical role in malignant hyperthermia crises, we treated CASQ1-null mice with two antioxidants, N-acetylcysteine (NAC, Sigma-Aldrich, Italy; provided ad libitum in drinking water) and (±)-6-hydroxy-2,5,7,8-tetramethylchromane-2-carboxylic acid (Trolox, Sigma-Aldrich; administered by intraperitoneal injection), before exposure to halothane (2%, 1 h) or heat (41°C, 1 h). RESULTS NAC and Trolox significantly protected CASQ1-null mice from lethal episodes, with mortality being 79% (n = 14), 25% (n = 16), and 20% (n = 5) during halothane exposure and 86% (n = 21), 29% (n = 21), and 33% (n = 6) during heat stress in untreated, NAC-treated, and Trolox-treated mice, respectively. During heat challenge, an increase in core temperature in CASQ1-null mice (42.3° ± 0.1°C, n=10) was significantly reduced by both NAC and Trolox (40.6° ± 0.3°C, n = 6 and 40.5° ± 0.2°C, n = 6). NAC treatment of CASQ1-null muscles/mice normalized caffeine sensitivity during in vitro contracture tests, Ca transients in single fibers, and significantly reduced the percentage of fibers undergoing rhabdomyolysis (37.6 ± 2.5%, 38/101 fibers in 3 mice; 11.6 ± 1.1%, 21/186 fibers in 5 mice). The protective effect of antioxidant treatment likely resulted from mitigation of oxidative stress, because NAC reduced mitochondrial superoxide production, superoxide dismutase type-1 expression, and 3-nitrotyrosine expression, and increased both reduced glutathione and reduced glutathione/oxidized glutathione ratio. CONCLUSION These studies provide a deeper understanding of the mechanisms that underlie hyperthermic crises in CASQ1-deficient muscle and demonstrate that antioxidant pretreatment may prevent them.
Collapse
Affiliation(s)
- Antonio Michelucci
- Postdoctoral Fellow, CeSI - Center for Research on Ageing & DNICS – Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti, I-66100 Chieti, Italy
| | - Cecilia Paolini
- Assistant Professor, CeSI - Center for Research on Ageing & DNICS – Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti, I-66100 Chieti, Italy
| | - Marta Canato
- Research Assistant, Department of Biomedical Sciences, University of Padova, I-35131 Italy
| | - Lan Wei-Lapierre
- Research Assistant Professor, Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Laura Pietrangelo
- Postdoctoral Fellow, CeSI - Center for Research on Ageing & DNICS – Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti, I-66100 Chieti, Italy
| | - Alessandro De Marco
- Postdoctoral fellow, CeSI - Center for Research on Ageing & DNICS – Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti, I-66100 Chieti, Italy
| | - Carlo Reggiani
- Professor, Department of Biomedical Sciences, University of Padova, I-35131 Italy
| | - Robert T. Dirksen
- Professor, Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642
| | - Feliciano Protasi
- Professor, CeSI - Center for Research on Ageing & DNICS – Department of Neuroscience, Imaging and Clinical Sciences, University G. d’Annunzio of Chieti, I-66100 Chieti, Italy
| |
Collapse
|