251
|
Rampelt H, Zerbes RM, van der Laan M, Pfanner N. Role of the mitochondrial contact site and cristae organizing system in membrane architecture and dynamics. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:737-746. [DOI: 10.1016/j.bbamcr.2016.05.020] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/12/2016] [Accepted: 05/17/2016] [Indexed: 12/22/2022]
|
252
|
Ong SB, Kalkhoran SB, Hernández-Reséndiz S, Samangouei P, Ong SG, Hausenloy DJ. Mitochondrial-Shaping Proteins in Cardiac Health and Disease - the Long and the Short of It! Cardiovasc Drugs Ther 2017; 31:87-107. [PMID: 28190190 PMCID: PMC5346600 DOI: 10.1007/s10557-016-6710-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondrial health is critically dependent on the ability of mitochondria to undergo changes in mitochondrial morphology, a process which is regulated by mitochondrial shaping proteins. Mitochondria undergo fission to generate fragmented discrete organelles, a process which is mediated by the mitochondrial fission proteins (Drp1, hFIS1, Mff and MiD49/51), and is required for cell division, and to remove damaged mitochondria by mitophagy. Mitochondria undergo fusion to form elongated interconnected networks, a process which is orchestrated by the mitochondrial fusion proteins (Mfn1, Mfn2 and OPA1), and which enables the replenishment of damaged mitochondrial DNA. In the adult heart, mitochondria are relatively static, are constrained in their movement, and are characteristically arranged into 3 distinct subpopulations based on their locality and function (subsarcolemmal, myofibrillar, and perinuclear). Although the mitochondria are arranged differently, emerging data supports a role for the mitochondrial shaping proteins in cardiac health and disease. Interestingly, in the adult heart, it appears that the pleiotropic effects of the mitochondrial fusion proteins, Mfn2 (endoplasmic reticulum-tethering, mitophagy) and OPA1 (cristae remodeling, regulation of apoptosis, and energy production) may play more important roles than their pro-fusion effects. In this review article, we provide an overview of the mitochondrial fusion and fission proteins in the adult heart, and highlight their roles as novel therapeutic targets for treating cardiac disease.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Siavash Beikoghli Kalkhoran
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Parisa Samangouei
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Derek John Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore. .,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore. .,The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK. .,The National Institute of Health Research, University College London Hospitals Biomedical Research Centre, London, UK.
| |
Collapse
|
253
|
Cardiolipin and mitochondrial cristae organization. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1859:1156-1163. [PMID: 28336315 DOI: 10.1016/j.bbamem.2017.03.013] [Citation(s) in RCA: 202] [Impact Index Per Article: 28.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2016] [Revised: 03/03/2017] [Accepted: 03/18/2017] [Indexed: 11/23/2022]
Abstract
A fundamental question in cell biology, under investigation for over six decades, is the structural organization of mitochondrial cristae. Long known to harbor electron transport chain proteins, crista membrane integrity is key to establishment of the proton gradient that drives oxidative phosphorylation. Visualization of cristae morphology by electron microscopy/tomography has provided evidence that cristae are tube-like extensions of the mitochondrial inner membrane (IM) that project into the matrix space. Reconciling ultrastructural data with the lipid composition of the IM provides support for a continuously curved cylindrical bilayer capped by a dome-shaped tip. Strain imposed by the degree of curvature is relieved by an asymmetric distribution of phospholipids in monolayer leaflets that comprise cristae membranes. The signature mitochondrial lipid, cardiolipin (~18% of IM phospholipid mass), and phosphatidylethanolamine (34%) segregate to the negatively curved monolayer leaflet facing the crista lumen while the opposing, positively curved, matrix-facing monolayer leaflet contains predominantly phosphatidylcholine. Associated with cristae are numerous proteins that function in distinctive ways to establish and/or maintain their lipid repertoire and structural integrity. By combining unique lipid components with a set of protein modulators, crista membranes adopt and maintain their characteristic morphological and functional properties. Once established, cristae ultrastructure has a direct impact on oxidative phosphorylation, apoptosis, fusion/fission as well as diseases of compromised energy metabolism.
Collapse
|
254
|
Bravo-Sagua R, Parra V, López-Crisosto C, Díaz P, Quest AFG, Lavandero S. Calcium Transport and Signaling in Mitochondria. Compr Physiol 2017; 7:623-634. [PMID: 28333383 DOI: 10.1002/cphy.c160013] [Citation(s) in RCA: 160] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Calcium (Ca2+) is a key player in the regulation of many cell functions. Just like Ca2+, mitochondria are ubiquitous, versatile, and dynamic players in determining both cell survival and death decisions. Given their ubiquitous nature, the regulation of both is deeply intertwined, whereby Ca2+ regulates mitochondrial functions, while mitochondria shape Ca2+ dynamics. Deregulation of either Ca2+ or mitochondrial signaling leads to abnormal function, cell damage or even cell death, thereby contributing to muscle dysfunction or cardiac pathologies. Moreover, altered mitochondrial Ca2+ homeostasis has been linked to metabolic diseases like cancer, obesity, and pulmonary hypertension. In this review article, we summarize the mechanisms that coordinate mitochondrial and Ca2+ responses and how they affect human health. © 2017 American Physiological Society. Compr Physiol 7:623-634, 2017.
Collapse
Affiliation(s)
- Roberto Bravo-Sagua
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.,Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Valentina Parra
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
| | - Camila López-Crisosto
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
| | - Paula Díaz
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile
| | - Andrew F G Quest
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Molecular Studies of the Cell (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences & Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Molecular Studies of the Cell (CEMC), Institute of Biomedical Sciences (ICBM), Faculty of Medicine, University of Chile, Santiago, Chile.,Cardiology Division, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
255
|
Lee H, Smith SB, Yoon Y. The short variant of the mitochondrial dynamin OPA1 maintains mitochondrial energetics and cristae structure. J Biol Chem 2017; 292:7115-7130. [PMID: 28298442 DOI: 10.1074/jbc.m116.762567] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 03/15/2017] [Indexed: 01/06/2023] Open
Abstract
The protein optic atrophy 1 (OPA1) is a dynamin-related protein associated with the inner mitochondrial membrane and functions in mitochondrial inner membrane fusion and cristae maintenance. Inner membrane-anchored long OPA1 (L-OPA1) undergoes proteolytic cleavage resulting in short OPA1 (S-OPA1). It is often thought that S-OPA1 is a functionally insignificant proteolytic product of L-OPA1 because the accumulation of S-OPA1 due to L-OPA1 cleavage is observed in mitochondrial fragmentation and dysfunction. However, cells contain a mixture of both L- and S-OPA1 in normal conditions, suggesting the functional significance of maintaining both OPA1 forms, but the differential roles of L- and S-OPA1 in mitochondrial fusion and energetics are ill-defined. Here, we examined mitochondrial fusion and energetic activities in cells possessing L-OPA1 alone, S-OPA1 alone, or both L- and S-OPA1. Using a mitochondrial fusion assay, we established that L-OPA1 confers fusion competence, whereas S-OPA1 does not. Remarkably, we found that S-OPA1 alone without L-OPA1 can maintain oxidative phosphorylation function as judged by growth in oxidative phosphorylation-requiring media, respiration measurements, and levels of the respiratory complexes. Most strikingly, S-OPA1 alone maintained normal mitochondrial cristae structure, which has been commonly assumed to be the function of OPA1 oligomers containing both L- and S-OPA1. Furthermore, we found that the GTPase activity of OPA1 is critical for maintaining cristae tightness and thus energetic competency. Our results demonstrate that, contrary to conventional notion, S-OPA1 is fully competent for maintaining mitochondrial energetics and cristae structure.
Collapse
Affiliation(s)
| | - Sylvia B Smith
- Cellular Biology and Anatomy, Medical College of Georgia, Augusta University, Augusta, Georgia 30912
| | | |
Collapse
|
256
|
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: 3.6] [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
|
257
|
Regulation of Mitochondrial Dynamics and Autophagy by the Mitochondria-Associated Membrane. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 997:33-47. [DOI: 10.1007/978-981-10-4567-7_3] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
258
|
Migdal I, Skibior-Blaszczyk R, Heidorn-Czarna M, Kolodziejczak M, Garbiec A, Janska H. AtOMA1 Affects the OXPHOS System and Plant Growth in Contrast to Other Newly Identified ATP-Independent Proteases in Arabidopsis Mitochondria. FRONTIERS IN PLANT SCIENCE 2017; 8:1543. [PMID: 28936218 PMCID: PMC5594102 DOI: 10.3389/fpls.2017.01543] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 08/23/2017] [Indexed: 05/17/2023]
Abstract
Compared with yeast, our knowledge on members of the ATP-independent plant mitochondrial proteolytic machinery is rather poor. In the present study, using confocal microscopy and immunoblotting, we proved that homologs of yeast Oma1, Atp23, Imp1, Imp2, and Oct1 proteases are localized in Arabidopsis mitochondria. We characterized these components of the ATP-independent proteolytic system as well as the earlier identified protease, AtICP55, with an emphasis on their significance in plant growth and functionality in the OXPHOS system. A functional complementation assay demonstrated that out of all the analyzed proteases, only AtOMA1 and AtICP55 could substitute for a lack of their yeast counterparts. We did not observe any significant developmental or morphological changes in plants lacking the studied proteases, either under optimal growth conditions or after exposure to stress, with the only exception being retarded root growth in oma1-1, thus implying that the absence of a single mitochondrial ATP-independent protease is not critical for Arabidopsis growth and development. We did not find any evidence indicating a clear functional complementation of the missing protease by any other protease at the transcript or protein level. Studies on the impact of the analyzed proteases on mitochondrial bioenergetic function revealed that out of all the studied mutants, only oma1-1 showed differences in activities and amounts of OXPHOS proteins. Among all the OXPHOS disorders found in oma1-1, the complex V deficiency is distinctive because it is mainly associated with decreased catalytic activity and not correlated with complex abundance, which has been observed in the case of supercomplex I + III2 and complex I deficiencies. Altogether, our study indicates that despite the presence of highly conservative homologs, the mitochondrial ATP-independent proteolytic system is not functionally conserved in plants as compared with yeast. Our findings also highlight the importance of AtOMA1 in maintenance of proper function of the OXPHOS system as well as in growth and development of Arabidopsis thaliana.
Collapse
Affiliation(s)
- Iwona Migdal
- Institute of Experimental Biology, Faculty of Biological Sciences, University of WroclawWroclaw, Poland
| | - Renata Skibior-Blaszczyk
- Department of Cellular Molecular Biology, Faculty of Biotechnology, University of WroclawWroclaw, Poland
| | - Malgorzata Heidorn-Czarna
- Department of Cellular Molecular Biology, Faculty of Biotechnology, University of WroclawWroclaw, Poland
| | - Marta Kolodziejczak
- Department of Cellular Molecular Biology, Faculty of Biotechnology, University of WroclawWroclaw, Poland
| | - Arnold Garbiec
- Institute of Experimental Biology, Faculty of Biological Sciences, University of WroclawWroclaw, Poland
| | - Hanna Janska
- Department of Cellular Molecular Biology, Faculty of Biotechnology, University of WroclawWroclaw, Poland
- *Correspondence: Hanna Janska,
| |
Collapse
|
259
|
Smith HL, Bourne JN, Cao G, Chirillo MA, Ostroff LE, Watson DJ, Harris KM. Mitochondrial support of persistent presynaptic vesicle mobilization with age-dependent synaptic growth after LTP. eLife 2016; 5. [PMID: 27991850 PMCID: PMC5235352 DOI: 10.7554/elife.15275] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2016] [Accepted: 12/16/2016] [Indexed: 12/22/2022] Open
Abstract
Mitochondria support synaptic transmission through production of ATP, sequestration of calcium, synthesis of glutamate, and other vital functions. Surprisingly, less than 50% of hippocampal CA1 presynaptic boutons contain mitochondria, raising the question of whether synapses without mitochondria can sustain changes in efficacy. To address this question, we analyzed synapses from postnatal day 15 (P15) and adult rat hippocampus that had undergone theta-burst stimulation to produce long-term potentiation (TBS-LTP) and compared them to control or no stimulation. At 30 and 120 min after TBS-LTP, vesicles were decreased only in presynaptic boutons that contained mitochondria at P15, and vesicle decrement was greatest in adult boutons containing mitochondria. Presynaptic mitochondrial cristae were widened, suggesting a sustained energy demand. Thus, mitochondrial proximity reflected enhanced vesicle mobilization well after potentiation reached asymptote, in parallel with the apparently silent addition of new dendritic spines at P15 or the silent enlargement of synapses in adults. DOI:http://dx.doi.org/10.7554/eLife.15275.001
Collapse
Affiliation(s)
- Heather L Smith
- Department of Neuroscience, Center for Learning and Memory, Institute for Neuroscience, University of Texas at Austin, Austin, United States
| | - Jennifer N Bourne
- Department of Cell and Developmental Biology, University of Colorado Denver - Anschutz Medical Campus, Aurora, United States
| | - Guan Cao
- Department of Neuroscience, Center for Learning and Memory, Institute for Neuroscience, University of Texas at Austin, Austin, United States
| | - Michael A Chirillo
- Department of Neuroscience, Center for Learning and Memory, Institute for Neuroscience, University of Texas at Austin, Austin, United States
| | - Linnaea E Ostroff
- Center for Neural Science, New York University, Washington, New York
| | - Deborah J Watson
- Department of Neuroscience, Center for Learning and Memory, Institute for Neuroscience, University of Texas at Austin, Austin, United States
| | - Kristen M Harris
- Department of Neuroscience, Center for Learning and Memory, Institute for Neuroscience, University of Texas at Austin, Austin, United States
| |
Collapse
|
260
|
Hesselink MKC, Schrauwen-Hinderling V, Schrauwen P. Skeletal muscle mitochondria as a target to prevent or treat type 2 diabetes mellitus. Nat Rev Endocrinol 2016; 12:633-645. [PMID: 27448057 DOI: 10.1038/nrendo.2016.104] [Citation(s) in RCA: 180] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Low levels of physical activity and the presence of obesity are associated with mitochondrial dysfunction. In addition, mitochondrial dysfunction has been associated with the development of insulin resistance and type 2 diabetes mellitus (T2DM). Although the evidence for a causal relationship between mitochondrial function and insulin resistance is still weak, emerging evidence indicates that boosting mitochondrial function might be beneficial to patient health. Exercise training is probably the most recognized promoter of mitochondrial function and insulin sensitivity and hence is still regarded as the best strategy to prevent and treat T2DM. Animal data, however, have revealed several new insights into the regulation of mitochondrial metabolism, and novel targets for interventions to boost mitochondrial function have emerged. Importantly, many of these targets seem to be regulated by factors such as nutrition, ambient temperature and circadian rhythms, which provides a basis for nonpharmacological strategies to prevent or treat T2DM in humans. Here, we will review the current evidence that mitochondrial function can be targeted therapeutically to improve insulin sensitivity and to prevent T2DM, focusing mainly on human intervention studies.
Collapse
Affiliation(s)
- Matthijs K C Hesselink
- Department of Human Biology and Human Movement Sciences, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
| | - Vera Schrauwen-Hinderling
- Department of Human Biology and Human Movement Sciences, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- Department of Radiology, Maastricht University Medical Center, P. Debyelaan 25, 6229 HX, Maastricht, Netherlands
| | - Patrick Schrauwen
- Department of Human Biology and Human Movement Sciences, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
- NUTRIM, School for Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, Universiteitsingel 50, 6229 ER, Maastricht, Netherlands
| |
Collapse
|
261
|
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: 167] [Impact Index Per Article: 20.9] [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
|
262
|
Delerue T, Khosrobakhsh F, Daloyau M, Emorine LJ, Dedieu A, Herbert CJ, Bonnefoy N, Arnauné-Pelloquin L, Belenguer P. Loss of Msp1p in Schizosaccharomyces pombe induces a ROS-dependent nuclear mutator phenotype that affects mitochondrial fission genes. FEBS Lett 2016; 590:3544-3558. [PMID: 27664110 DOI: 10.1002/1873-3468.12432] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 06/29/2016] [Accepted: 08/31/2016] [Indexed: 01/12/2023]
Abstract
Mitochondria continually fuse and divide to dynamically adapt to changes in metabolism and stress. Mitochondrial dynamics are also required for mitochondrial DNA (mtDNA) integrity; however, the underlying reason is not known. In this study, we examined the link between mitochondrial fusion and mtDNA maintenance in Schizosaccharomyces pombe, which cannot survive without mtDNA, by screening for suppressors of the lethality induced by loss of the dynamin-related large GTPase Msp1p. Our findings reveal that inactivation of Msp1p induces a ROS-dependent nuclear mutator phenotype that affects mitochondrial fission genes involved in suppressing mitochondrial fragmentation and mtDNA depletion. This indicates that mitochondrial fusion is crucial for maintaining the integrity of both mitochondrial and nuclear genetic information. Furthermore, our study suggests that the primary roles of Msp1p are to organize mitochondrial membranes, thus making them competent for fusion, and maintain the integrity of mtDNA.
Collapse
Affiliation(s)
- Thomas Delerue
- Center of Developmental Biology (CBD) and Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI), Toulouse University, CNRS, UPS, France
| | - Farnoosh Khosrobakhsh
- Center of Developmental Biology (CBD) and Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI), Toulouse University, CNRS, UPS, France.,Department of Biological Science, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Marlène Daloyau
- Center of Developmental Biology (CBD) and Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI), Toulouse University, CNRS, UPS, France
| | - Laurent Jean Emorine
- Center of Developmental Biology (CBD) and Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI), Toulouse University, CNRS, UPS, France
| | - Adrien Dedieu
- Center of Developmental Biology (CBD) and Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI), Toulouse University, CNRS, UPS, France
| | - Christopher J Herbert
- Institute of Integrative Biology of the Cell (I2BC), CEA, CNRS, University Paris-Sud, University Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Nathalie Bonnefoy
- Institute of Integrative Biology of the Cell (I2BC), CEA, CNRS, University Paris-Sud, University Paris-Saclay, Gif-sur-Yvette Cedex, France
| | - Laetitia Arnauné-Pelloquin
- Center of Developmental Biology (CBD) and Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI), Toulouse University, CNRS, UPS, France
| | - Pascale Belenguer
- Center of Developmental Biology (CBD) and Research Center on Animal Cognition (CRCA), Center for Integrative Biology (CBI), Toulouse University, CNRS, UPS, France.
| |
Collapse
|
263
|
Horibata Y, Ando H, Zhang P, Vergnes L, Aoyama C, Itoh M, Reue K, Sugimoto H. StarD7 Protein Deficiency Adversely Affects the Phosphatidylcholine Composition, Respiratory Activity, and Cristae Structure of Mitochondria. J Biol Chem 2016; 291:24880-24891. [PMID: 27694445 DOI: 10.1074/jbc.m116.736793] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Revised: 08/30/2016] [Indexed: 12/17/2022] Open
Abstract
Phosphatidylcholine (PC) is a major phospholipid of mitochondria, comprising 40-50% of both the outer and the inner membranes. However, PC must be imported from its production organelles because mitochondria lack the enzymes essential for PC biosynthesis. In a previous study, we found that StarD7 mediates the intracellular transfer of PC to mitochondria. Therefore, in this study, we analyzed the contribution of StarD7 to the maintenance of mitochondrial phospholipid content and function using siRNA-mediated knockdown and knock-out (KO) of the StarD7 gene in HEPA-1 cells. Real time analysis of respiratory activity demonstrated that the oxygen consumption rate and activity of mitochondrial complexes were impaired in StarD7-KD cells. To confirm these results, we established StarD7-KO HEPA-1 cells by double nicking using CRISPR/Cas9n. As expected, StarD7-KD and -KO cells showed a significant reduction in mitochondrial PC content. The ATP level and growth rate of KO cells were notably lower compared with wild-type cells when cultured in glucose-free galactose-containing medium to force cells to rely on mitochondrial ATP production. In KO cells, the level of the MTCO1 protein, a primary subunit of complex IV, was reduced without a concomitant decrease in its mRNA, but the level was restored when StarD7-I was overexpressed. StarD7-KO cells showed impaired formation of the mitochondrial supercomplexes and exhibited a disorganized cristae structure, with no changes in optic atrophy 1 protein. These findings indicate that StarD7 plays important roles in maintaining the proper composition of mitochondrial phospholipids as well as mitochondrial function and morphogenesis.
Collapse
Affiliation(s)
- Yasuhiro Horibata
- From the Department of Biochemistry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, Japan and
| | - Hiromi Ando
- From the Department of Biochemistry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, Japan and
| | - Peixiang Zhang
- the Department of Human Genetics, David Geffen School of Medicine and
| | - Laurent Vergnes
- the Department of Human Genetics, David Geffen School of Medicine and
| | - Chieko Aoyama
- From the Department of Biochemistry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, Japan and
| | - Masahiko Itoh
- From the Department of Biochemistry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, Japan and
| | - Karen Reue
- the Department of Human Genetics, David Geffen School of Medicine and.,the Molecular Biology Institute at UCLA, Los Angeles, California 90095
| | - Hiroyuki Sugimoto
- From the Department of Biochemistry, Dokkyo Medical University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi, Japan and
| |
Collapse
|
264
|
Barrera M, Koob S, Dikov D, Vogel F, Reichert AS. OPA1 functionally interacts with MIC60 but is dispensable for crista junction formation. FEBS Lett 2016; 590:3309-3322. [DOI: 10.1002/1873-3468.12384] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 07/13/2016] [Accepted: 07/13/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Miguel Barrera
- Mitochondrial Biology; Buchmann Institute of Molecular Life Sciences; Goethe University Frankfurt; Frankfurt am Main Germany
| | - Sebastian Koob
- Mitochondrial Biology; Buchmann Institute of Molecular Life Sciences; Goethe University Frankfurt; Frankfurt am Main Germany
| | - Daniel Dikov
- Mitochondrial Biology; Buchmann Institute of Molecular Life Sciences; Goethe University Frankfurt; Frankfurt am Main Germany
| | - Frank Vogel
- Max-Delbrück-Centrum für Molekulare Medizin; Berlin/Buch Germany
| | - Andreas S. Reichert
- Mitochondrial Biology; Buchmann Institute of Molecular Life Sciences; Goethe University Frankfurt; Frankfurt am Main Germany
- Institute of Biochemistry and Molecular Biology I; Medical Faculty; Heinrich-Heine University Düsseldorf; Germany
| |
Collapse
|
265
|
Plecitá-Hlavatá L, Ježek P. Integration of superoxide formation and cristae morphology for mitochondrial redox signaling. Int J Biochem Cell Biol 2016; 80:31-50. [PMID: 27640755 DOI: 10.1016/j.biocel.2016.09.010] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 09/09/2016] [Accepted: 09/12/2016] [Indexed: 12/16/2022]
Abstract
The mitochondrial network provides the central cell's energetic and regulatory unit, which besides ATP and metabolite production participates in cellular signaling through regulated reactive oxygen species (ROS) production and various protein/ion fluxes. The inner membrane forms extensive folds, called cristae, i.e. cavities enfolded from and situated perpendicularly to its inner boundary membrane portion, which encompasses an inner cylinder within the outer membrane tubule. Mitochondrial cristae ultramorphology reflects various metabolic, physiological or pathological states. Since the mitochondrion is typically a predominant superoxide source and generated ROS also serve for the creation of information redox signals, we review known relationships between ROS generation within the respiratory chain complexes of cristae and cristae morphology. Notably, it is emphasized that cristae shape is governed by ATP-synthase dimers, MICOS complexes, OPA1 isoforms and the umbrella of their regulation, and also dependent on local protonmotive force (electrical potential component) in cristae. Cristae are also affected by redox-sensitive kinases/phosphatases or p66SHC. ATP-synthase dimers decrease in the inflated intracristal space, diminishing pH and hypothetically having minimal superoxide formation. Matrix-released signaling superoxide/H2O2 is predominantly integrated along mitochondrial tubules, whereas the diffusion of intracristal signaling ROS species is controlled by crista junctions, the widening of which enables specific retrograde redox signaling such as during hypoxic cell adaptation. Other physiological cases of H2O2 release from the mitochondrion include the modulation of insulin release in pancreatic β-cells, enhancement of insulin signaling in peripheral tissues, signaling by T-cell receptors, retrograde signaling during the cell cycle and cell differentiation, specifically that of adipocytes.
Collapse
Affiliation(s)
- Lydie Plecitá-Hlavatá
- Department of Membrane Transport Biophysics, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Petr Ježek
- Department of Membrane Transport Biophysics, No.75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic.
| |
Collapse
|
266
|
Janer A, Prudent J, Paupe V, Fahiminiya S, Majewski J, Sgarioto N, Des Rosiers C, Forest A, Lin ZY, Gingras AC, Mitchell G, McBride HM, Shoubridge EA. SLC25A46 is required for mitochondrial lipid homeostasis and cristae maintenance and is responsible for Leigh syndrome. EMBO Mol Med 2016; 8:1019-38. [PMID: 27390132 PMCID: PMC5009808 DOI: 10.15252/emmm.201506159] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Mitochondria form a dynamic network that responds to physiological signals and metabolic stresses by altering the balance between fusion and fission. Mitochondrial fusion is orchestrated by conserved GTPases MFN1/2 and OPA1, a process coordinated in yeast by Ugo1, a mitochondrial metabolite carrier family protein. We uncovered a homozygous missense mutation in SLC25A46, the mammalian orthologue of Ugo1, in a subject with Leigh syndrome. SLC25A46 is an integral outer membrane protein that interacts with MFN2, OPA1, and the mitochondrial contact site and cristae organizing system (MICOS) complex. The subject mutation destabilizes the protein, leading to mitochondrial hyperfusion, alterations in endoplasmic reticulum (ER) morphology, impaired cellular respiration, and premature cellular senescence. The MICOS complex is disrupted in subject fibroblasts, resulting in strikingly abnormal mitochondrial architecture, with markedly shortened cristae. SLC25A46 also interacts with the ER membrane protein complex EMC, and phospholipid composition is altered in subject mitochondria. These results show that SLC25A46 plays a role in a mitochondrial/ER pathway that facilitates lipid transfer, and link altered mitochondrial dynamics to early‐onset neurodegenerative disease and cell fate decisions.
Collapse
Affiliation(s)
- Alexandre Janer
- Department of Human Genetics, McGill University, Montreal, QC, Canada Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Julien Prudent
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Vincent Paupe
- Department of Human Genetics, McGill University, Montreal, QC, Canada Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | | | - Jacek Majewski
- Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Nicolas Sgarioto
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| | - Christine Des Rosiers
- Department of Nutrition, Université de Montréal, Montreal, QC, Canada Research Centre, Montreal Heart Institute, Montreal, QC, Canada
| | - Anik Forest
- Department of Nutrition, Université de Montréal, Montreal, QC, Canada Research Centre, Montreal Heart Institute, Montreal, QC, Canada
| | - Zhen-Yuan Lin
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, Canada Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Grant Mitchell
- Division of Medical Genetics, Department of Pediatrics, CHU Sainte-Justine and Université de Montréal, Montreal, QC, Canada
| | - Heidi M McBride
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada
| | - Eric A Shoubridge
- Department of Human Genetics, McGill University, Montreal, QC, Canada Montreal Neurological Institute, McGill University, Montreal, QC, Canada
| |
Collapse
|
267
|
Korwitz A, Merkwirth C, Richter-Dennerlein R, Tröder SE, Sprenger HG, Quirós PM, López-Otín C, Rugarli EI, Langer T. Loss of OMA1 delays neurodegeneration by preventing stress-induced OPA1 processing in mitochondria. J Cell Biol 2016; 212:157-66. [PMID: 26783299 PMCID: PMC4738383 DOI: 10.1083/jcb.201507022] [Citation(s) in RCA: 100] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Loss of OMA1 in a mouse model of neurodegeneration stabilizes fusion-active L-OPA1, which supports neuronal survival by preventing apoptosis, independent of its effects on cristae morphogenesis. Proteolytic cleavage of the dynamin-like guanosine triphosphatase OPA1 in mitochondria is emerging as a central regulatory hub that determines mitochondrial morphology under stress and in disease. Stress-induced OPA1 processing by OMA1 triggersmitochondrial fragmentation, which is associated with mitophagy and apoptosis in vitro. Here, we identify OMA1 as a critical regulator of neuronal survival in vivo and demonstrate that stress-induced OPA1 processing by OMA1 promotes neuronal death and neuroinflammatory responses. Using mice lacking prohibitin membrane scaffolds as a model of neurodegeneration, we demonstrate that additional ablation of Oma1 delays neuronal loss and prolongs lifespan. This is accompanied by the accumulation of fusion-active, long OPA1 forms, which stabilize the mitochondrial genome but do not preserve mitochondrial cristae or respiratory chain supercomplex assembly in prohibitin-depleted neurons. Thus, long OPA1 forms can promote neuronal survival independently of cristae shape, whereas stress-induced OMA1 activation and OPA1 cleavage limit mitochondrial fusion and promote neuronal death.
Collapse
Affiliation(s)
- Anne Korwitz
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50931 Cologne, Germany
| | - Carsten Merkwirth
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50931 Cologne, Germany
| | - Ricarda Richter-Dennerlein
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50931 Cologne, Germany
| | - Simon E Tröder
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50931 Cologne, Germany
| | - Hans-Georg Sprenger
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50931 Cologne, Germany
| | - Pedro M Quirós
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo, Oviedo 33006, Spain
| | - Carlos López-Otín
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Universitario de Oncología, Universidad de Oviedo, Oviedo 33006, Spain
| | - Elena I Rugarli
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50931 Cologne, Germany Center for Molecular Medicine, University of Cologne, 50931 Cologne, Germany
| | - Thomas Langer
- Institute of Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, 50931 Cologne, Germany Center for Molecular Medicine, University of Cologne, 50931 Cologne, Germany Max Planck Institute for Biology of Aging, 50931 Cologne, Germany
| |
Collapse
|
268
|
El-Sikhry HE, Alsaleh N, Dakarapu R, Falck JR, Seubert JM. Novel Roles of Epoxyeicosanoids in Regulating Cardiac Mitochondria. PLoS One 2016; 11:e0160380. [PMID: 27494529 PMCID: PMC4975494 DOI: 10.1371/journal.pone.0160380] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2016] [Accepted: 07/18/2016] [Indexed: 11/18/2022] Open
Abstract
Maintenance of a healthy pool of mitochondria is important for the function and survival of terminally differentiated cells such as cardiomyocytes. Epoxyeicosatrienoic acids (EETs) are epoxy lipids derived from metabolism of arachidonic acid by cytochrome P450 epoxygenases. We have previously shown that EETs trigger a protective response limiting mitochondrial dysfunction and reducing cellular death. The aim of this study was to investigate whether EET-mediated effects influence mitochondrial quality in HL-1 cardiac cells during starvation. HL-1 cells were subjected to serum- and amino acid free conditions for 24h. We employed a dual-acting synthetic analog UA-8 (13-(3-propylureido)tridec-8-enoic acid), possessing both EET-mimetic and soluble epoxide hydrolase (sEH) inhibitory properties, or 14,15-EET as model EET molecules. We demonstrated that EET-mediated events significantly improved mitochondrial function as assessed by preservation of the ADP/ATP ratio and oxidative respiratory capacity. Starvation induced mitochondrial hyperfusion observed in control cells was attenuated by UA-8. However, EET-mediated events did not affect the expression of mitochondrial dynamic proteins Fis1, DRP-1 or Mfn2. Rather we observed increased levels of OPA-1 oligomers and increased mitochondrial cristae density, which correlated with the preserved mitochondrial function. Increased DNA binding activity of pCREB and Nrf1/2 and increased SIRT1 activity together with elevated mitochondrial proteins suggest EET-mediated events led to preserved mitobiogenesis. Thus, we provide new evidence for EET-mediated events that preserve a healthier pool of mitochondria in cardiac cells following starvation-induced stress.
Collapse
Affiliation(s)
- Haitham E. El-Sikhry
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Nasser Alsaleh
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
| | - Rambabu Dakarapu
- Departments of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - John R. Falck
- Departments of Biochemistry and Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, United States of America
| | - John M. Seubert
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Pharmacology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
- * E-mail:
| |
Collapse
|
269
|
Bioenergetic roles of mitochondrial fusion. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:1277-1283. [DOI: 10.1016/j.bbabio.2016.04.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 03/18/2016] [Accepted: 04/05/2016] [Indexed: 11/17/2022]
|
270
|
García-Bermúdez J, Cuezva JM. The ATPase Inhibitory Factor 1 (IF1): A master regulator of energy metabolism and of cell survival. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1857:1167-1182. [PMID: 26876430 DOI: 10.1016/j.bbabio.2016.02.004] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/04/2015] [Revised: 01/28/2016] [Accepted: 02/07/2016] [Indexed: 12/19/2022]
Abstract
In this contribution we summarize most of the findings reported for the molecular and cellular biology of the physiological inhibitor of the mitochondrial H(+)-ATP synthase, the engine of oxidative phosphorylation (OXPHOS) and gate of cell death. We first describe the structure and major mechanisms and molecules that regulate the activity of the ATP synthase placing the ATPase Inhibitory Factor 1 (IF1) as a major determinant in the regulation of the activity of the ATP synthase and hence of OXPHOS. Next, we summarize the post-transcriptional mechanisms that regulate the expression of IF1 and emphasize, in addition to the regulation afforded by the protonation state of histidine residues, that the activity of IF1 as an inhibitor of the ATP synthase is also regulated by phosphorylation of a serine residue. Phosphorylation of S39 in IF1 by the action of a mitochondrial cAMP-dependent protein kinase A hampers its interaction with the ATP synthase, i.e., only dephosphorylated IF1 interacts with the enzyme. Upon IF1 interaction with the ATP synthase both the synthetic and hydrolytic activities of the engine of OXPHOS are inhibited. These findings are further placed into the physiological context to stress the emerging roles played by IF1 in metabolic reprogramming in cancer, in hypoxia and in cellular differentiation. We review also the implication of IF1 in other cellular situations that involve the malfunctioning of mitochondria. Special emphasis is given to the role of IF1 as driver of the generation of a reactive oxygen species signal that, emanating from mitochondria, is able to reprogram the nucleus of the cell to confer by various signaling pathways a cell-death resistant phenotype against oxidative stress. Overall, our intention is to highlight the urgent need of further investigations in the molecular and cellular biology of IF1 and of its target, the ATP synthase, to unveil new therapeutic strategies in human pathology. This article is part of a Special Issue entitled 'EBEC 2016: 19th European Bioenergetics Conference, Riva del Garda, Italy, July 2-6, 2016', edited by Prof. Paolo Bernardi.
Collapse
Affiliation(s)
- Javier García-Bermúdez
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras CIBERER-ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, 28049, Madrid, Spain.
| |
Collapse
|
271
|
van der Laan M, Horvath SE, Pfanner N. Mitochondrial contact site and cristae organizing system. Curr Opin Cell Biol 2016; 41:33-42. [DOI: 10.1016/j.ceb.2016.03.013] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2016] [Revised: 03/19/2016] [Accepted: 03/23/2016] [Indexed: 10/22/2022]
|
272
|
Kushnareva Y, Seong Y, Andreyev AY, Kuwana T, Kiosses WB, Votruba M, Newmeyer DD. Mitochondrial dysfunction in an Opa1(Q285STOP) mouse model of dominant optic atrophy results from Opa1 haploinsufficiency. Cell Death Dis 2016; 7:e2309. [PMID: 27468686 PMCID: PMC4973340 DOI: 10.1038/cddis.2016.160] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 04/29/2016] [Accepted: 05/02/2016] [Indexed: 12/22/2022]
Abstract
Mutations in the opa1 (optic atrophy 1) gene lead to autosomal dominant optic atrophy (ADOA), a hereditary eye disease. This gene encodes the Opa1 protein, a mitochondrial dynamin-related GTPase required for mitochondrial fusion and the maintenance of normal crista structure. The majority of opa1 mutations encode truncated forms of the protein, lacking a complete GTPase domain. It is unclear whether the phenotype results from haploinsufficiency or rather a deleterious effect of truncated Opa1 protein. We studied a heterozygous Opa1 mutant mouse carrying a defective allele with a stop codon in the beginning of the GTPase domain at residue 285, a mutation that mimics human pathological mutations. Using an antibody raised against an N-terminal portion of Opa1, we found that the level of wild-type protein was decreased in the mutant mice, as predicted. However, no truncated Opa1 protein was expressed. In embryonic fibroblasts isolated from the mutant mice, this partial loss of Opa1 caused mitochondrial respiratory deficiency and a selective loss of respiratory Complex IV subunits. Furthermore, partial Opa1 deficiency resulted in a substantial resistance to endoplasmic reticulum stress-induced death. On the other hand, the enforced expression of truncated Opa1 protein in cells containing normal levels of wild-type protein did not cause mitochondrial defects. Moreover, cells expressing the truncated Opa1 protein showed reduced Bax activation in response to apoptotic stimuli. Taken together, our results exclude deleterious dominant-negative or gain-of-function mechanisms for this type of Opa1 mutation and affirm haploinsufficiency as the mechanism underlying mitochondrial dysfunction in ADOA.
Collapse
Affiliation(s)
- Y Kushnareva
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Y Seong
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - A Y Andreyev
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | - T Kuwana
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - W B Kiosses
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - M Votruba
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4LU, UK.,Cardiff Eye Unit, University Hospital Wales, Cardiff CF14 4XW, UK
| | - D D Newmeyer
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| |
Collapse
|
273
|
Buck MD, O'Sullivan D, Klein Geltink RI, Curtis JD, Chang CH, Sanin DE, Qiu J, Kretz O, Braas D, van der Windt GJW, Chen Q, Huang SCC, O'Neill CM, Edelson BT, Pearce EJ, Sesaki H, Huber TB, Rambold AS, Pearce EL. Mitochondrial Dynamics Controls T Cell Fate through Metabolic Programming. Cell 2016; 166:63-76. [PMID: 27293185 DOI: 10.1016/j.cell.2016.05.035] [Citation(s) in RCA: 968] [Impact Index Per Article: 121.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2015] [Revised: 04/03/2016] [Accepted: 05/06/2016] [Indexed: 01/07/2023]
Abstract
Activated effector T (TE) cells augment anabolic pathways of metabolism, such as aerobic glycolysis, while memory T (TM) cells engage catabolic pathways, like fatty acid oxidation (FAO). However, signals that drive these differences remain unclear. Mitochondria are metabolic organelles that actively transform their ultrastructure. Therefore, we questioned whether mitochondrial dynamics controls T cell metabolism. We show that TE cells have punctate mitochondria, while TM cells maintain fused networks. The fusion protein Opa1 is required for TM, but not TE cells after infection, and enforcing fusion in TE cells imposes TM cell characteristics and enhances antitumor function. Our data suggest that, by altering cristae morphology, fusion in TM cells configures electron transport chain (ETC) complex associations favoring oxidative phosphorylation (OXPHOS) and FAO, while fission in TE cells leads to cristae expansion, reducing ETC efficiency and promoting aerobic glycolysis. Thus, mitochondrial remodeling is a signaling mechanism that instructs T cell metabolic programming.
Collapse
Affiliation(s)
- Michael D Buck
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David O'Sullivan
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Ramon I Klein Geltink
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Jonathan D Curtis
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Chih-Hao Chang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David E Sanin
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Jing Qiu
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Oliver Kretz
- Renal Division, University Medical Center Freiburg, 79106 Freiburg, Germany; Department of Neuroanatomy, University of Freiburg, 79104 Freiburg, Germany
| | - Daniel Braas
- University of California Los Angeles Metabolomics Center, Los Angeles, CA 90095, USA
| | | | - Qiongyu Chen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Stanley Ching-Cheng Huang
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Christina M O'Neill
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Brian T Edelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Edward J Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany; Faculty of Biology, University of Freiburg, 79104 Freiburg, Germany
| | - Hiromi Sesaki
- Department of Cell Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Tobias B Huber
- Renal Division, University Medical Center Freiburg, 79106 Freiburg, Germany; BIOSS Centre for Biological Signaling Studies, 79104 Freiburg, Germany
| | - Angelika S Rambold
- Center for Chronic Immunodeficiency, University Medical Center Freiburg and University of Freiburg, 79106 Freiburg, Germany; Department of Developmental Immunology, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Erika L Pearce
- Department of Immunometabolism, Max Planck Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany.
| |
Collapse
|
274
|
MacVicar T, Langer T. OPA1 processing in cell death and disease - the long and short of it. J Cell Sci 2016; 129:2297-306. [PMID: 27189080 DOI: 10.1242/jcs.159186] [Citation(s) in RCA: 256] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The regulation of mitochondrial dynamics by the GTPase OPA1, which is located at the inner mitochondrial membrane, is crucial for adapting mitochondrial function and preserving cellular health. OPA1 governs the delicate balance between fusion and fission in the dynamic mitochondrial network. A disturbance of this balance, often observed under stress and pathologic conditions, causes mitochondrial fragmentation and can ultimately result in cell death. As discussed in this Commentary, these morphological changes are regulated by proteolytic processing of OPA1 by the inner-membrane peptidases YME1L (also known as YME1L1) and OMA1. Long, membrane-bound forms of OPA1 are required for mitochondrial fusion, but their processing to short, soluble forms limits fusion and can facilitate mitochondrial fission. Excessive OPA1 processing by the stress-activated protease OMA1 promotes mitochondrial fragmentation and, if persistent, triggers cell death and tissue degeneration in vivo The prevention of OMA1-mediated OPA1 processing and mitochondrial fragmentation might thus offer exciting therapeutic potential for human diseases associated with mitochondrial dysfunction.
Collapse
Affiliation(s)
- Thomas MacVicar
- Institute of Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| | - Thomas Langer
- Institute of Genetics, Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD) and Center for Molecular Medicine (CMMC), University of Cologne, Cologne 50931, Germany
| |
Collapse
|
275
|
The Chromatin Remodeling Complex Chd4/NuRD Controls Striated Muscle Identity and Metabolic Homeostasis. Cell Metab 2016; 23:881-92. [PMID: 27166947 DOI: 10.1016/j.cmet.2016.04.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Revised: 02/19/2016] [Accepted: 04/13/2016] [Indexed: 01/01/2023]
Abstract
Heart muscle maintains blood circulation, while skeletal muscle powers skeletal movement. Despite having similar myofibrilar sarcomeric structures, these striated muscles differentially express specific sarcomere components to meet their distinct contractile requirements. The mechanism responsible is still unclear. We show here that preservation of the identity of the two striated muscle types depends on epigenetic repression of the alternate lineage gene program by the chromatin remodeling complex Chd4/NuRD. Loss of Chd4 in the heart triggers aberrant expression of the skeletal muscle program, causing severe cardiomyopathy and sudden death. Conversely, genetic depletion of Chd4 in skeletal muscle causes inappropriate expression of cardiac genes and myopathy. In both striated tissues, mitochondrial function was also dependent on the Chd4/NuRD complex. We conclude that an epigenetic mechanism controls cardiac and skeletal muscle structural and metabolic identities and that loss of this regulation leads to hybrid striated muscle tissues incompatible with life.
Collapse
|
276
|
Demers-Lamarche J, Guillebaud G, Tlili M, Todkar K, Bélanger N, Grondin M, Nguyen AP, Michel J, Germain M. Loss of Mitochondrial Function Impairs Lysosomes. J Biol Chem 2016; 291:10263-76. [PMID: 26987902 PMCID: PMC4858975 DOI: 10.1074/jbc.m115.695825] [Citation(s) in RCA: 151] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 03/04/2016] [Indexed: 02/04/2023] Open
Abstract
Alterations in mitochondrial function, as observed in neurodegenerative diseases, lead to disrupted energy metabolism and production of damaging reactive oxygen species. Here, we demonstrate that mitochondrial dysfunction also disrupts the structure and function of lysosomes, the main degradation and recycling organelle. Specifically, inhibition of mitochondrial function, following deletion of the mitochondrial protein AIF, OPA1, or PINK1, as well as chemical inhibition of the electron transport chain, impaired lysosomal activity and caused the appearance of large lysosomal vacuoles. Importantly, our results show that lysosomal impairment is dependent on reactive oxygen species. Given that alterations in both mitochondrial function and lysosomal activity are key features of neurodegenerative diseases, this work provides important insights into the etiology of neurodegenerative diseases.
Collapse
Affiliation(s)
- Julie Demers-Lamarche
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and Centre de recherche Biomed, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada and
| | - Gérald Guillebaud
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and Centre de recherche Biomed, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada and
| | - Mouna Tlili
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and
| | - Kiran Todkar
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and Centre de recherche Biomed, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada and
| | - Noémie Bélanger
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and
| | - Martine Grondin
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and
| | - Angela P Nguyen
- the Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Jennifer Michel
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and
| | - Marc Germain
- From the Groupe de Recherche en Signalisation Cellulaire, Département de Biologie Médicale and Centre de recherche Biomed, Université du Québec à Trois-Rivières, Trois-Rivières, Québec G9A 5H7, Canada and
| |
Collapse
|
277
|
Rainbolt TK, Lebeau J, Puchades C, Wiseman RL. Reciprocal Degradation of YME1L and OMA1 Adapts Mitochondrial Proteolytic Activity during Stress. Cell Rep 2016; 14:2041-2049. [PMID: 26923599 DOI: 10.1016/j.celrep.2016.02.011] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2015] [Revised: 12/21/2015] [Accepted: 01/29/2016] [Indexed: 12/31/2022] Open
Abstract
The mitochondrial inner membrane proteases YME1L and OMA1 are critical regulators of essential mitochondrial functions, including inner membrane proteostasis maintenance and mitochondrial dynamics. Here, we show that YME1L and OMA1 are reciprocally degraded in response to distinct types of cellular stress. OMA1 is degraded through a YME1L-dependent mechanism in response to toxic insults that depolarize the mitochondrial membrane. Alternatively, insults that depolarize mitochondria and deplete cellular ATP stabilize active OMA1 and promote YME1L degradation. We show that the differential degradation of YME1L and OMA1 alters their proteolytic processing of the dynamin-like GTPase OPA1, a critical regulator of mitochondrial inner membrane morphology, which influences the recovery of tubular mitochondria following membrane-depolarization-induced fragmentation. Our results reveal the differential stress-induced degradation of YME1L and OMA1 as a mechanism for sensitively adapting mitochondrial inner membrane protease activity and function in response to distinct types of cellular insults.
Collapse
Affiliation(s)
- T Kelly Rainbolt
- Department of Molecular and Experimental Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Justine Lebeau
- Department of Molecular and Experimental Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - Cristina Puchades
- Department of Molecular and Experimental Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | - R Luke Wiseman
- Department of Molecular and Experimental Medicine, Department of Chemical Physiology, The Scripps Research Institute, La Jolla, CA 92037, USA.
| |
Collapse
|
278
|
Otera H, Miyata N, Kuge O, Mihara K. Drp1-dependent mitochondrial fission via MiD49/51 is essential for apoptotic cristae remodeling. J Cell Biol 2016; 212:531-44. [PMID: 26903540 PMCID: PMC4772499 DOI: 10.1083/jcb.201508099] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 01/26/2016] [Indexed: 11/25/2022] Open
Abstract
Drp1-dependent mitochondrial fission through MiD49 and MiD51, but not through Mff, is essential for cristae remodeling to facilitate cytochrome c release during the early phase of intrinsic apoptosis. Mitochondrial fission facilitates cytochrome c release from the intracristae space into the cytoplasm during intrinsic apoptosis, although how the mitochondrial fission factor Drp1 and its mitochondrial receptors Mff, MiD49, and MiD51 are involved in this reaction remains elusive. Here, we analyzed the functional division of these receptors with their knockout (KO) cell lines. In marked contrast to Mff-KO cells, MiD49/MiD51-KO and Drp1-KO cells completely resisted cristae remodeling and cytochrome c release during apoptosis. This phenotype in MiD49/51-KO cells, but not Drp1-KO cells, was completely abolished by treatments disrupting cristae structure such as OPA1 depletion. Unexpectedly, OPA1 oligomers generally thought to resist cytochrome c release by stabilizing the cristae structure were similarly disassembled in Drp1-KO and MiD49/51-KO cells, indicating that disassembly of OPA1 oligomers is not directly linked to cristae remodeling for cytochrome c release. Together, these results indicate that Drp1-dependent mitochondrial fission through MiD49/MiD51 regulates cristae remodeling during intrinsic apoptosis.
Collapse
Affiliation(s)
- Hidenori Otera
- Graduate School of Medical Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Non Miyata
- Department of Chemistry, Faculty of Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Osamu Kuge
- Department of Chemistry, Faculty of Sciences, Kyushu University, Fukuoka 819-0395, Japan
| | - Katsuyoshi Mihara
- Graduate School of Medical Sciences, Kyushu University, Fukuoka 819-0395, Japan
| |
Collapse
|
279
|
Plecitá-Hlavatá L, Engstová H, Alán L, Špaček T, Dlasková A, Smolková K, Špačková J, Tauber J, Strádalová V, Malínský J, Lessard M, Bewersdorf J, Ježek P. Hypoxic HepG2 cell adaptation decreases ATP synthase dimers and ATP production in inflated cristae by mitofilin down-regulation concomitant to MICOS clustering. FASEB J 2016; 30:1941-57. [PMID: 26887443 DOI: 10.1096/fj.201500176] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2015] [Accepted: 01/24/2016] [Indexed: 01/25/2023]
Abstract
The relationship of the inner mitochondrial membrane (IMM) cristae structure and intracristal space (ICS) to oxidative phosphorylation (oxphos) is not well understood. Mitofilin (subunit Mic60) of the mitochondrial contact site and cristae organizing system (MICOS) IMM complex is attached to the outer membrane (OMM) via the sorting and assembly machinery/topogenesis of mitochondrial outer membrane β-barrel proteins (SAM/TOB) complex and controls the shape of the cristae. ATP synthase dimers determine sharp cristae edges, whereas trimeric OPA1 tightens ICS outlets. Metabolism is altered during hypoxia, and we therefore studied cristae morphology in HepG2 cells adapted to 5% oxygen for 72 h. Three dimensional (3D), super-resolution biplane fluorescence photoactivation localization microscopy with Eos-conjugated, ICS-located lactamase-β indicated hypoxic ICS expansion with an unchanged OMM (visualized by Eos-mitochondrial fission protein-1). 3D direct stochastic optical reconstruction microscopy immunocytochemistry revealed foci of clustered mitofilin (but not MICOS subunit Mic19) in contrast to its even normoxic distribution. Mitofilin mRNA and protein decreased by ∼20%. ATP synthase dimers vs monomers and state-3/state-4 respiration ratios were lower during hypoxia. Electron microscopy confirmed ICS expansion (maximum in glycolytic cells), which was absent in reduced or OMM-detached cristae of OPA1- and mitofilin-silenced cells, respectively. Hypoxic adaptation is reported as rounding sharp cristae edges and expanding cristae width (ICS) by partial mitofilin/Mic60 down-regulation. Mitofilin-depleted MICOS detaches from SAM while remaining MICOS with mitofilin redistributes toward higher interdistances. This phenomenon causes partial oxphos dormancy in glycolytic cells via disruption of ATP synthase dimers.-Plecitá-Hlavatá, L., Engstová, H., Alán, L., Špaček, T., Dlasková, A., Smolková, K., Špačková, J., Tauber, J., Strádalová, V., Malínský, J., Lessard, M., Bewersdorf, J., Ježek, P. Hypoxic HepG2 cell adaptation decreases ATP synthase dimers and ATP production in inflated cristae by mitofilin down-regulation concomitant to MICOS clustering.
Collapse
Affiliation(s)
- Lydie Plecitá-Hlavatá
- Department of Membrane Transport Biophysics, Department No. 75, Institute of Physiology, and
| | - Hana Engstová
- Department of Membrane Transport Biophysics, Department No. 75, Institute of Physiology, and
| | - Lukáš Alán
- Department of Membrane Transport Biophysics, Department No. 75, Institute of Physiology, and
| | - Tomáš Špaček
- Department of Membrane Transport Biophysics, Department No. 75, Institute of Physiology, and
| | - Andrea Dlasková
- Department of Membrane Transport Biophysics, Department No. 75, Institute of Physiology, and
| | - Katarína Smolková
- Department of Membrane Transport Biophysics, Department No. 75, Institute of Physiology, and
| | - Jitka Špačková
- Department of Membrane Transport Biophysics, Department No. 75, Institute of Physiology, and
| | - Jan Tauber
- Department of Membrane Transport Biophysics, Department No. 75, Institute of Physiology, and
| | - Vendula Strádalová
- Microscopy Unit, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Jan Malínský
- Microscopy Unit, Institute of Experimental Medicine, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Mark Lessard
- Institute for Molecular Biophysics, The Jackson Laboratory, Bar Harbor, Maine, USA; and
| | - Joerg Bewersdorf
- Department of Cell Biology, Yale University, New Haven, Connecticut, USA
| | - Petr Ježek
- Department of Membrane Transport Biophysics, Department No. 75, Institute of Physiology, and
| |
Collapse
|
280
|
Wai T, García-Prieto J, Baker MJ, Merkwirth C, Benit P, Rustin P, Rupérez FJ, Barbas C, Ibañez B, Langer T. Imbalanced OPA1 processing and mitochondrial fragmentation cause heart failure in mice. Science 2016; 350:aad0116. [PMID: 26785494 DOI: 10.1126/science.aad0116] [Citation(s) in RCA: 377] [Impact Index Per Article: 47.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Mitochondrial morphology is shaped by fusion and division of their membranes. Here, we found that adult myocardial function depends on balanced mitochondrial fusion and fission, maintained by processing of the dynamin-like guanosine triphosphatase OPA1 by the mitochondrial peptidases YME1L and OMA1. Cardiac-specific ablation of Yme1l in mice activated OMA1 and accelerated OPA1 proteolysis, which triggered mitochondrial fragmentation and altered cardiac metabolism. This caused dilated cardiomyopathy and heart failure. Cardiac function and mitochondrial morphology were rescued by Oma1 deletion, which prevented OPA1 cleavage. Feeding mice a high-fat diet or ablating Yme1l in skeletal muscle restored cardiac metabolism and preserved heart function without suppressing mitochondrial fragmentation. Thus, unprocessed OPA1 is sufficient to maintain heart function, OMA1 is a critical regulator of cardiomyocyte survival, and mitochondrial morphology and cardiac metabolism are intimately linked.
Collapse
Affiliation(s)
- Timothy Wai
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany. Max-Planck-Institute for Biology of Aging, Cologne, Germany
| | - Jaime García-Prieto
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain
| | - Michael J Baker
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
| | - Carsten Merkwirth
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
| | - Paule Benit
- INSERM UMR 1141, Hôpital Robert Debré, Paris, France. Université Paris 7, Faculté de Médecine Denis Diderot, Paris, France
| | - Pierre Rustin
- INSERM UMR 1141, Hôpital Robert Debré, Paris, France. Université Paris 7, Faculté de Médecine Denis Diderot, Paris, France
| | - Francisco Javier Rupérez
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo CEU, Campus Monteprincipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Coral Barbas
- Centre for Metabolomics and Bioanalysis (CEMBIO), Faculty of Pharmacy, Universidad San Pablo CEU, Campus Monteprincipe, Boadilla del Monte, 28668 Madrid, Spain
| | - Borja Ibañez
- Myocardial Pathophysiology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain. Department of Cardiology, Instituto de Investigación Sanitaria (IIS), Fundación Jiménez Díaz Hospital, Madrid, Spain.
| | - Thomas Langer
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany. Max-Planck-Institute for Biology of Aging, Cologne, Germany. Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany. Center for Molecular Medicine (CMMC), University of Cologne, Cologne, Germany.
| |
Collapse
|
281
|
A randomized, placebo-controlled trial of the benzoquinone idebenone in a mouse model of OPA1-related dominant optic atrophy reveals a limited therapeutic effect on retinal ganglion cell dendropathy and visual function. Neuroscience 2016; 319:92-106. [PMID: 26820596 DOI: 10.1016/j.neuroscience.2016.01.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 12/19/2022]
Abstract
Dominant optic atrophy (DOA) arises from mutations in the OPA1 gene that promotes fusion of the inner mitochondrial membrane and plays a role in maintaining ATP levels. Patients display optic disc pallor, retinal ganglion cell (RGC) loss and bilaterally reduced vision. We report a randomized, placebo-controlled trial of idebenone at 2000 mg/kg/day in 56 Opa1 mutant mice (B6;C3-Opa1(Q285STOP)), with RGC dendropathy and visual loss, and 63 wildtype mice. We assessed cellular responses in the retina, brain and liver and RGC morphology, by diolistic labeling, Sholl analysis and quantification of dendritic morphometric features. Vision was assessed by optokinetic responses. ATP levels were raised by 0.57 nmol/mg (97.73%, p=0.035) in brain from idebenone-treated Opa1 mutant mice, but in the liver there was an 80.35% (p=0.011) increase in oxidative damage. NQO1 expression in Opa1 mutant mice was reduced in the brain (to 30.5%, p=0.002) but not in retina, and neither expression level was induced by idebenone. ON-center RGCs failed to show major recovery, other than improvements in secondary dendritic length (by 53.89%, p=0.052) and dendritic territory (by 2.22 × 10(4) μm(2) or 90.24%, p=0.074). An improvement in optokinetic response was observed (by 12.2 ± 3.2s, p=0.003), but this effect was not sustained over time. OFF-center RGCs from idebenone-treated wildtype mice showed shrinkage in total dendritic length by 2.40 mm (48.05%, p=0.025) and a 47.37% diminished Sholl profile (p=0.029). Visual function in wildtype idebenone-treated mice was impaired (2.9 fewer head turns than placebo, p=0.007). Idebenone appears largely ineffective in protecting Opa1 heterozygous RGCs from dendropathy. The detrimental effect of idebenone in wildtype mice has not been previously observed and raises some concerns.
Collapse
|
282
|
|
283
|
Abstract
Mitochondrial dynamics, fission and fusion, were first identified in yeast with investigation in heart cells beginning only in the last 5 to 7 years. In the ensuing time, it has become evident that these processes are not only required for healthy mitochondria, but also, that derangement of these processes contributes to disease. The fission and fusion proteins have a number of functions beyond the mitochondrial dynamics. Many of these functions are related to their membrane activities, such as apoptosis. However, other functions involve other areas of the mitochondria, such as OPA1's role in maintaining cristae structure and preventing cytochrome c leak, and its essential (at least a 10 kDa fragment of OPA1) role in mtDNA replication. In heart disease, changes in expression of these important proteins can have detrimental effects on mitochondrial and cellular function.
Collapse
Affiliation(s)
- A A Knowlton
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| | - T T Liu
- Molecular & Cellular Cardiology, Division of Cardiovascular Medicine and Pharmacology Department, University of California, Davis, and The Department of Veteran's Affairs, Northern California VA, Sacramento, California, USA
| |
Collapse
|
284
|
Pernas L, Scorrano L. Mito-Morphosis: Mitochondrial Fusion, Fission, and Cristae Remodeling as Key Mediators of Cellular Function. Annu Rev Physiol 2015; 78:505-31. [PMID: 26667075 DOI: 10.1146/annurev-physiol-021115-105011] [Citation(s) in RCA: 521] [Impact Index Per Article: 57.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Permanent residency in the eukaryotic cell pressured the prokaryotic mitochondrial ancestor to strategize for intracellular living. Mitochondria are able to autonomously integrate and respond to cellular cues and demands by remodeling their morphology. These processes define mitochondrial dynamics and inextricably link the fate of the mitochondrion and that of the host eukaryote, as exemplified by the human diseases that result from mutations in mitochondrial dynamics proteins. In this review, we delineate the architecture of mitochondria and define the mechanisms by which they modify their shape. Key players in these mechanisms are discussed, along with their role in manipulating mitochondrial morphology during cellular action and development. Throughout, we highlight the evolutionary context in which mitochondrial dynamics emerged and consider unanswered questions whose dissection might lead to mitochondrial morphology-based therapies.
Collapse
Affiliation(s)
- Lena Pernas
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy; ,
| | - Luca Scorrano
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine, 35129 Padova, Italy; ,
| |
Collapse
|
285
|
Magalon A, Alberge F. Distribution and dynamics of OXPHOS complexes in the bacterial cytoplasmic membrane. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2015; 1857:198-213. [PMID: 26545610 DOI: 10.1016/j.bbabio.2015.10.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 12/23/2022]
Abstract
Oxidative phosphorylation (OXPHOS) is an essential process for most living organisms mostly sustained by protein complexes embedded in the cell membrane. In order to thrive, cells need to quickly respond to changes in the metabolic demand or in their environment. An overview of the strategies that can be employed by bacterial cells to adjust the OXPHOS outcome is provided. Regulation at the level of gene expression can only provide a means to adjust the OXPHOS outcome to long-term trends in the environment. In addition, the actual view is that bioenergetic membranes are highly compartmentalized structures. This review discusses what is known about the spatial organization of OXPHOS complexes and the timescales at which they occur. As exemplified with the commensal gut bacterium Escherichia coli, three levels of spatial organization are at play: supercomplexes, membrane microdomains and polar assemblies. This review provides a particular focus on whether dynamic spatial organization can fine-tune the OXPHOS through the definition of specialized functional membrane microdomains. Putative mechanisms responsible for spatio-temporal regulation of the OXPHOS complexes are discussed. This article is part of a Special Issue entitled Organization and dynamics of bioenergetic systems in bacteria, edited by Conrad Mullineaux.
Collapse
Affiliation(s)
- Axel Magalon
- CNRS, Laboratoire de Chimie Bactérienne (UMR 7283), Institut de Microbiologie de la Méditerranée, 13009 Marseille, France; Aix-Marseille University, UMR 7283, 13009 Marseille, France.
| | - François Alberge
- CNRS, Laboratoire de Chimie Bactérienne (UMR 7283), Institut de Microbiologie de la Méditerranée, 13009 Marseille, France; Aix-Marseille University, UMR 7283, 13009 Marseille, France
| |
Collapse
|
286
|
Sun Y, Xue W, Song Z, Huang K, Zheng L. Restoration of Opa1-long isoform inhibits retinal injury-induced neurodegeneration. J Mol Med (Berl) 2015; 94:335-46. [PMID: 26530815 DOI: 10.1007/s00109-015-1359-y] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 10/19/2015] [Accepted: 10/22/2015] [Indexed: 12/21/2022]
Abstract
Optic atrophy 1 (Opa1) is a critical factor that regulates fusion and other important functions of mitochondria. In mitochondrion, the N-terminal mitochondrial targeting sequence of Opa1 precursors is removed to generate Opa1 long isoforms (L-Opa1), which are further cleaved into short isoforms (S-Opa1). In the present study, we found that retinal ischemia-reperfusion (I/R) injury and intravitreal injection of carbonylcyanide m-chlorophenyl hydrazone (CCCP) both dramatically induced Opa1 cleavage and caused loss of L-Opa1. In cultured neuronal cells under hypoxia-reoxygenation (H/R) injury, similar changes for Opa1 were also observed. In contrast, restoration of L-Opa1 level by overexpression of S1 cleavage site deletion Opa1 splice 1 (Opa1-ΔS1) not only normalized the H/R-induced mitochondrial morphology changes, but also inhibited the H/R-induced apoptosis, necrosis, and the intracellular ATP loss. Furthermore, recovering L-Opa1 level in the I/R-injured retina by intravitreal injection of genipin or overexpression of Opa1-ΔS1 inhibited apoptosis, necrosis, cell loss in the ganglion cell layer and retinal thickness reduction. Together, our data demonstrated the loss of L-Opa1 is involved in the development of retinal I/R injury, indicating restoring L-Opa1 level may be considered as a therapeutic target for I/R injury-related diseases, at least for the retina. Key messages: Retinal ischemia-reperfusion (I/R) or hypoxia-reoxygenation (H/R) injury induces L-Opa1 loss. Opa1-ΔS1 overexpression inhibits H/R-induced L-Opa1 loss. Opa1-ΔS1 overexpression inhibits H/R-induced mitochondria morphology change. Opa1-ΔS1 and genipin inhibit retinal I/R injury-induced necroptosis. Opa1-ΔS1 and genipin inhibit retinal I/R injury-induced neurodegeneration.
Collapse
Affiliation(s)
- Yue Sun
- College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, People's Republic of China
| | - Weili Xue
- College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, People's Republic of China
| | - Zhiyin Song
- College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, People's Republic of China
| | - Kun Huang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, People's Republic of China.
| | - Ling Zheng
- College of Life Sciences, Wuhan University, Wuhan, Hubei, 430072, People's Republic of China.
| |
Collapse
|
287
|
Bertholet AM, Delerue T, Millet AM, Moulis MF, David C, Daloyau M, Arnauné-Pelloquin L, Davezac N, Mils V, Miquel MC, Rojo M, Belenguer P. Mitochondrial fusion/fission dynamics in neurodegeneration and neuronal plasticity. Neurobiol Dis 2015; 90:3-19. [PMID: 26494254 DOI: 10.1016/j.nbd.2015.10.011] [Citation(s) in RCA: 255] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/16/2015] [Accepted: 10/13/2015] [Indexed: 12/17/2022] Open
Abstract
Mitochondria are dynamic organelles that continually move, fuse and divide. The dynamic balance of fusion and fission of mitochondria determines their morphology and allows their immediate adaptation to energetic needs, keeps mitochondria in good health by restoring or removing damaged organelles or precipitates cells in apoptosis in cases of severe defects. Mitochondrial fusion and fission are essential in mammals and their disturbances are associated with several diseases. However, while mitochondrial fusion/fission dynamics, and the proteins that control these processes, are ubiquitous, associated diseases are primarily neurological disorders. Accordingly, inactivation of the main actors of mitochondrial fusion/fission dynamics is associated with defects in neuronal development, plasticity and functioning, both ex vivo and in vivo. Here, we present the central actors of mitochondrial fusion and fission and review the role of mitochondrial dynamics in neuronal physiology and pathophysiology. Particular emphasis is placed on the three main actors of these processes i.e. DRP1,MFN1-2, and OPA1 as well as on GDAP1, a protein of the mitochondrial outer membrane preferentially expressed in neurons. This article is part of a Special Issue entitled: Mitochondria & Brain.
Collapse
Affiliation(s)
- A M Bertholet
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - T Delerue
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - A M Millet
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - M F Moulis
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - C David
- CNRS, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France
| | - M Daloyau
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - L Arnauné-Pelloquin
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - N Davezac
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - V Mils
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - M C Miquel
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - M Rojo
- CNRS, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France.
| | - P Belenguer
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France.
| |
Collapse
|
288
|
Abstract
Mitochondrial dysfunction underlies many human disorders, including those that affect the visual system. The retinal ganglion cells, whose axons form the optic nerve, are often damaged by mitochondrial-related diseases which result in blindness. Both mitochondrial DNA (mtDNA) and nuclear gene mutations impacting many different mitochondrial processes can result in optic nerve disease. Of particular importance are mutations that impair mitochondrial network dynamics (fusion and fission), oxidative phosphorylation (OXPHOS), and formation of iron-sulfur complexes. Current genetic knowledge can inform genetic counseling and suggest strategies for novel gene-based therapies. Identifying new optic neuropathy-causing genes and defining the role of current and novel genes in disease will be important steps toward the development of effective and potentially neuroprotective therapies.
Collapse
Affiliation(s)
- Janey L Wiggs
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, Massachusetts 02114;
| |
Collapse
|
289
|
Lavatelli F, Imperlini E, Orrù S, Rognoni P, Sarnataro D, Palladini G, Malpasso G, Soriano ME, Di Fonzo A, Valentini V, Gnecchi M, Perlini S, Salvatore F, Merlini G. Novel mitochondrial protein interactors of immunoglobulin light chains causing heart amyloidosis. FASEB J 2015. [PMID: 26220173 DOI: 10.1096/fj.15-272179] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In immunoglobulin (Ig) light-chain (LC) (AL) amyloidosis, AL deposition translates into life-threatening cardiomyopathy. Clinical and experimental evidence indicates that soluble cardiotoxic LCs are themselves harmful for cells, by which they are internalized. Hypothesizing that interaction of soluble cardiotoxic LCs with cellular proteins contributes to damage, we characterized their interactome in cardiac cells. LCs were purified from patients with AL amyloidosis cardiomyopathy or multiple myeloma without amyloidosis (the nonamyloidogenic/noncardiotoxic LCs served as controls) and employed at concentrations in the range observed in AL patients' sera. A functional proteomic approach, based on direct and inverse coimmunoprecipitation and mass spectrometry, allowed identifying LC-protein complexes. Findings were validated by colocalization, fluorescence lifetime imaging microscopy (FLIM)-fluorescence resonance energy transfer (FRET), and ultrastructural studies, using human primary cardiac fibroblasts (hCFs) and stem cell-derived cardiomyocytes. Amyloidogenic cardiotoxic LCs interact in vitro with specific intracellular proteins involved in viability and metabolism. Imaging confirmed that, especially in hCFs, cardiotoxic LCs (not controls) colocalize with mitochondria and spatially associate with selected interactors: mitochondrial optic atrophy 1-like protein and peroxisomal acyl-coenzyme A oxidase 1 (FLIM-FRET efficiencies 11 and 6%, respectively). Cardiotoxic LC-treated hCFs display mitochondrial ultrastructural changes, supporting mitochondrial involvement. We show that cardiotoxic LCs establish nonphysiologic protein-protein contacts in human cardiac cells, offering new clues on the pathogenesis of AL cardiomyopathy.
Collapse
Affiliation(s)
- Francesca Lavatelli
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Esther Imperlini
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Stefania Orrù
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Paola Rognoni
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Daniela Sarnataro
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Giuseppina Palladini
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Giuseppe Malpasso
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Maria Eugenia Soriano
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Andrea Di Fonzo
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Veronica Valentini
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Massimiliano Gnecchi
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Stefano Perlini
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Francesco Salvatore
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| | - Giampaolo Merlini
- *Amyloidosis Research and Treatment Center, Department of Molecular Medicine, **Department of Internal Medicine, Department of Cardiothoracic and Vascular Sciences, Laboratory of Experimental Cardiology for Cell and Molecular Therapy, University of Pavia, and Clinical Chemistry Laboratory, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy; Centro di Ricerca di Ingegneria Genetica (CEINGE)-Biotecnologie Avanzate, Naples, Italy; Department of Movement Sciences, Parthenope University of Naples, Naples, Italy; Department of Molecular Medicine and Medical Biotechnologies, University of Naples Federico II, Naples, Italy; Department of Biology, University of Padua, Padua, Italy; and Department of Medicine, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
290
|
A novel OPA1 mutation causing variable age of onset autosomal dominant optic atrophy plus in an Australian family. J Neurol 2015; 262:2323-8. [PMID: 26194196 DOI: 10.1007/s00415-015-7849-6] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Revised: 07/06/2015] [Accepted: 07/06/2015] [Indexed: 10/23/2022]
Abstract
Pathogenic mutations in the OPA1 gene can be associated with Autosomal Dominant Optic Atrophy (ADOA). In approximately 20 % of patients with OPA1 mutations, a more complex neurodegenerative disorder with extraocular manifestations, known as ADOA Plus, can arise. 12 members of a multigenerational family were assessed clinically and screened for a genetic mutation in OPA1. Eight family members displayed manifestations consistent with ADOA Plus and four did not. Affected members of the oldest available generation displayed the most severe phenotype, which included severe optic atrophy, deafness, ptosis, ophthalmoplegia, proximal myopathy, neuropathy and ataxia. The next generation was less severely affected but several members displayed manifestations only after the fifth decade. Genetic analysis revealed a heterozygous variant in the OPA1 gene (c.1053T>A, p.Asp351Glu) that segregated with disease. The affected family members described here exhibited visual loss later than is typical for OPA1-related disease, as well as later onset of other neurological abnormalities in the fifth or sixth decades of life that progressed to severe neurological disability by the seventh decade. These findings expand the clinical spectrum of OPA1-related disease associated with a novel OPA1 mutation.
Collapse
|
291
|
Influence of Glucose Deprivation on Membrane Potentials of Plasma Membranes, Mitochondria and Synaptic Vesicles in Rat Brain Synaptosomes. Neurochem Res 2015; 40:1188-96. [DOI: 10.1007/s11064-015-1579-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Revised: 04/02/2015] [Accepted: 04/08/2015] [Indexed: 12/26/2022]
|
292
|
Hoitzing H, Johnston IG, Jones NS. What is the function of mitochondrial networks? A theoretical assessment of hypotheses and proposal for future research. Bioessays 2015; 37:687-700. [PMID: 25847815 PMCID: PMC4672710 DOI: 10.1002/bies.201400188] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Mitochondria can change their shape from discrete isolated organelles to a large continuous reticulum. The cellular advantages underlying these fused networks are still incompletely understood. In this paper, we describe and compare hypotheses regarding the function of mitochondrial networks. We use mathematical and physical tools both to investigate existing hypotheses and to generate new ones, and we suggest experimental and modelling strategies. Among the novel insights we underline from this work are the possibilities that (i) selective mitophagy is not required for quality control because selective fusion is sufficient; (ii) increased connectivity may have non-linear effects on the diffusion rate of proteins; and (iii) fused networks can act to dampen biochemical fluctuations. We hope to convey to the reader that quantitative approaches can drive advances in the understanding of the physiological advantage of these morphological changes.
Collapse
Affiliation(s)
- Hanne Hoitzing
- Department of Mathematics, Imperial College London, London, UK
| | - Iain G Johnston
- Department of Mathematics, Imperial College London, London, UK
| | - Nick S Jones
- Department of Mathematics, Imperial College London, London, UK
| |
Collapse
|
293
|
Suresh HG, da Silveira Dos Santos AX, Kukulski W, Tyedmers J, Riezman H, Bukau B, Mogk A. Prolonged starvation drives reversible sequestration of lipid biosynthetic enzymes and organelle reorganization in Saccharomyces cerevisiae. Mol Biol Cell 2015; 26:1601-15. [PMID: 25761633 PMCID: PMC4436773 DOI: 10.1091/mbc.e14-11-1559] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2014] [Accepted: 03/02/2015] [Indexed: 11/11/2022] Open
Abstract
Lipid homeostasis is modulated upon starvation at three different levels manifested in reversible 1) spatial confinement of lipid biosynthetic enzymes, 2) mitochondrial and endoplasmic reticular reorganization, and 3) loss of organelle contact sites, thus highlighting a novel mechanism regulating lipid biosynthesis by simply modulating flux through the pathway. Cells adapt to changing nutrient availability by modulating a variety of processes, including the spatial sequestration of enzymes, the physiological significance of which remains controversial. These enzyme deposits are claimed to represent aggregates of misfolded proteins, protein storage, or complexes with superior enzymatic activity. We monitored spatial distribution of lipid biosynthetic enzymes upon glucose depletion in Saccharomyces cerevisiae. Several different cytosolic-, endoplasmic reticulum–, and mitochondria-localized lipid biosynthetic enzymes sequester into distinct foci. Using the key enzyme fatty acid synthetase (FAS) as a model, we show that FAS foci represent active enzyme assemblies. Upon starvation, phospholipid synthesis remains active, although with some alterations, implying that other foci-forming lipid biosynthetic enzymes might retain activity as well. Thus sequestration may restrict enzymes' access to one another and their substrates, modulating metabolic flux. Enzyme sequestrations coincide with reversible drastic mitochondrial reorganization and concomitant loss of endoplasmic reticulum–mitochondria encounter structures and vacuole and mitochondria patch organelle contact sites that are reflected in qualitative and quantitative changes in phospholipid profiles. This highlights a novel mechanism that regulates lipid homeostasis without profoundly affecting the activity status of involved enzymes such that, upon entry into favorable growth conditions, cells can quickly alter lipid flux by relocalizing their enzymes.
Collapse
Affiliation(s)
- Harsha Garadi Suresh
- Center for Molecular Biology of the University of Heidelberg (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | | | - Wanda Kukulski
- Structural and Computational Biology Unit and Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany Structural and Computational Biology Unit and Cell Biology and Biophysics Unit, European Molecular Biology Laboratory, D-69117 Heidelberg, Germany
| | - Jens Tyedmers
- Department of Medicine I and Clinical Chemistry, University of Heidelberg, D-69120 Heidelberg, Germany
| | - Howard Riezman
- NCCR Chemical Biology, Department of Biochemistry, University of Geneva, CH-1211 Geneva, Switzerland
| | - Bernd Bukau
- Center for Molecular Biology of the University of Heidelberg (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Axel Mogk
- Center for Molecular Biology of the University of Heidelberg (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| |
Collapse
|
294
|
C11orf83, a mitochondrial cardiolipin-binding protein involved in bc1 complex assembly and supercomplex stabilization. Mol Cell Biol 2015; 35:1139-56. [PMID: 25605331 DOI: 10.1128/mcb.01047-14] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Mammalian mitochondria may contain up to 1,500 different proteins, and many of them have neither been confidently identified nor characterized. In this study, we demonstrated that C11orf83, which was lacking experimental characterization, is a mitochondrial inner membrane protein facing the intermembrane space. This protein is specifically associated with the bc1 complex of the electron transport chain and involved in the early stages of its assembly by stabilizing the bc1 core complex. C11orf83 displays some overlapping functions with Cbp4p, a yeast bc1 complex assembly factor. Therefore, we suggest that C11orf83, now called UQCC3, is the functional human equivalent of Cbp4p. In addition, C11orf83 depletion in HeLa cells caused abnormal crista morphology, higher sensitivity to apoptosis, a decreased ATP level due to impaired respiration and subtle, but significant, changes in cardiolipin composition. We showed that C11orf83 binds to cardiolipin by its α-helices 2 and 3 and is involved in the stabilization of bc1 complex-containing supercomplexes, especially the III2/IV supercomplex. We also demonstrated that the OMA1 metalloprotease cleaves C11orf83 in response to mitochondrial depolarization, suggesting a role in the selection of cells with damaged mitochondria for their subsequent elimination by apoptosis, as previously described for OPA1.
Collapse
|
295
|
Germain M. OPA1 and mitochondrial solute carriers in bioenergetic metabolism. Mol Cell Oncol 2015; 2:e982378. [PMID: 27308447 DOI: 10.4161/23723556.2014.982378] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Revised: 10/27/2014] [Accepted: 10/28/2014] [Indexed: 11/19/2022]
Abstract
The mitochondrial fusion protein optic atrophy 1 (OPA1) is required to maintain cristae structure and for ATP synthase assembly. Our recent work demonstrates that OPA1 dynamically regulates these processes by sensing changes in nutrient availability through mitochondrial solute carriers and adjusting the metabolic output of mitochondria accordingly. This is a critical survival process as its inhibition leads to cell death.
Collapse
Affiliation(s)
- Marc Germain
- Groupe de Recherche en Neurosciences; Département de Biologie Médicale; Université du Québec à Trois-Rivières ; Trois-Rivières, Canada
| |
Collapse
|
296
|
Khacho M, S. Slack R. Mitochondrial dynamics in neurodegeneration: from cell death to energetic states. AIMS MOLECULAR SCIENCE 2015. [DOI: 10.3934/molsci.2015.2.161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
297
|
Abstract
Recent studies link changes in energy metabolism with the fate of pluripotent stem cells (PSCs). Safe use of PSC derivatives in regenerative medicine requires an enhanced understanding and control of factors that optimize in vitro reprogramming and differentiation protocols. Relative shifts in metabolism from naïve through "primed" pluripotent states to lineage-directed differentiation place variable demands on mitochondrial biogenesis and function for cell types with distinct energetic and biosynthetic requirements. In this context, mitochondrial respiration, network dynamics, TCA cycle function, and turnover all have the potential to influence reprogramming and differentiation outcomes. Shifts in cellular metabolism affect enzymes that control epigenetic configuration, which impacts chromatin reorganization and gene expression changes during reprogramming and differentiation. Induced PSCs (iPSCs) may have utility for modeling metabolic diseases caused by mutations in mitochondrial DNA, for which few disease models exist. Here, we explore key features of PSC energy metabolism research in mice and man and the impact this work is starting to have on our understanding of early development, disease modeling, and potential therapeutic applications.
Collapse
Affiliation(s)
- Tara Teslaa
- Molecular Biology Institute, University of California, Los Angeles, CA, USA
| | - Michael A Teitell
- Molecular Biology Institute, University of California, Los Angeles, CA, USA Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA Department of Bioengineering, University of California, Los Angeles, CA, USA Department of Pediatrics, University of California, Los Angeles, CA, USA California NanoSystems Institute, University of California, Los Angeles, CA, USA Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, USA Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, CA, USA
| |
Collapse
|