1
|
Ravindran S, Rau CD. The multifaceted role of mitochondria in cardiac function: insights and approaches. Cell Commun Signal 2024; 22:525. [PMID: 39472951 PMCID: PMC11523909 DOI: 10.1186/s12964-024-01899-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/19/2024] [Indexed: 11/02/2024] Open
Abstract
Cardiovascular disease (CVD) remains a global economic burden even in the 21st century with 85% of deaths resulting from heart attacks. Despite efforts in reducing the risk factors, and enhancing pharmacotherapeutic strategies, challenges persist in early identification of disease progression and functional recovery of damaged hearts. Targeting mitochondrial dysfunction, a key player in the pathogenesis of CVD has been less successful due to its role in other coexisting diseases. Additionally, it is the only organelle with an agathokakological function that is a remedy and a poison for the cell. In this review, we describe the origins of cardiac mitochondria and the role of heteroplasmy and mitochondrial subpopulations namely the interfibrillar, subsarcolemmal, perinuclear, and intranuclear mitochondria in maintaining cardiac function and in disease-associated remodeling. The cumulative evidence of mitochondrial retrograde communication with the nucleus is addressed, highlighting the need to study the genotype-phenotype relationships of specific organelle functions with CVD by using approaches like genome-wide association study (GWAS). Finally, we discuss the practicality of computational methods combined with single-cell sequencing technologies to address the challenges of genetic screening in the identification of heteroplasmy and contributory genes towards CVD.
Collapse
Affiliation(s)
- Sriram Ravindran
- Computational Medicine Program, Department of Genetics, and McAllister Heart Institute, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC-27599, USA
| | - Christoph D Rau
- Computational Medicine Program, Department of Genetics, and McAllister Heart Institute, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC-27599, USA.
| |
Collapse
|
2
|
Kolitsida P, Saha A, Caliri A, Assali E, Riera AM, Itskanov S, Magana CS, Stork B, Shirihai O, Sekler I, Koehler CM, van der Bliek AM. Mfn2 induces NCLX-mediated calcium release from mitochondria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.05.606704. [PMID: 39149365 PMCID: PMC11326197 DOI: 10.1101/2024.08.05.606704] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/17/2024]
Abstract
Mfn2 is a mitochondrial outer membrane fusion protein with the additional role of tethering mitochondria to the ER. Here, we describe a novel connection between Mfn2 and calcium release from mitochondria. We show that Mfn2 controls the mitochondrial inner membrane sodium-calcium exchange protein NCLX, which is a major source for calcium release from mitochondria. This discovery was made with the fungal toxin Phomoxanthone (PXA), which induces calcium release from mitochondria. PXA-induced calcium release is blocked by a chemical inhibitor of NCLX, while NCLX and Mfn2 deletions both also prevent PXA-induced calcium release. CETSA experiments show that PXA directly targets Mfn2, which likely controls NCLX through physical interactions since co-immunoprecipitation and proximity ligation assays show increased association between Mfn2 and NCLX upon treatment with PXA. Interactions between Mfn2 and NCLX also increase when cells are treated with mitochondrial ROS-inducing conditions, such as oligomycin treatment of respiring cells, while the interactions do not increase in Oma1 -/- cells. It seems likely that opening of cristae by Oma1-mediated cleavage of Opa1 promotes translocation of NCLX from cristae to the rim where it can come into contact with Mfn2 thus promoting PXA-induced calcium release from mitochondria. These results therefore delineate a pathway that connects ROS produced inside mitochondria with calcium release and signaling in the cytosol.
Collapse
Affiliation(s)
| | - Akash Saha
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
| | - Andrew Caliri
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
| | - Essam Assali
- Department of Physiology and Cell Biology, Ben Gurion University, Israel
- Current: Yale School of Medicine, New haven CT
| | - Alejandro Martorell Riera
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
- Current: Kite Pharmaceutical, Santa Monica CA
| | - Samuel Itskanov
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
- Current: Gilead Sciences, San Francisco CA
| | - Catalina S Magana
- Department of Biological Chemistry, David Geffen School of Medicine at UCLA
| | - Björn Stork
- Institute of Molecular Medicine I, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Germany
| | - Orian Shirihai
- Department of Medicine, David Geffen School of Medicine at UCLA
| | - Israel Sekler
- Department of Physiology and Cell Biology, Ben Gurion University, Israel
| | | | | |
Collapse
|
3
|
Li H, Dai X, Zhou J, Wang Y, Zhang S, Guo J, Shen L, Yan H, Jiang H. Mitochondrial dynamics in pulmonary disease: Implications for the potential therapeutics. J Cell Physiol 2024:e31370. [PMID: 38988059 DOI: 10.1002/jcp.31370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 06/18/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
Mitochondria are dynamic organelles that continuously undergo fusion/fission to maintain normal cell physiological activities and energy metabolism. When mitochondrial dynamics is unbalanced, mitochondrial homeostasis is broken, thus damaging mitochondrial function. Accumulating evidence demonstrates that impairment in mitochondrial dynamics leads to lung tissue injury and pulmonary disease progression in a variety of disease models, including inflammatory responses, apoptosis, and barrier breakdown, and that the role of mitochondrial dynamics varies among pulmonary diseases. These findings suggest that modulation of mitochondrial dynamics may be considered as a valid therapeutic strategy in pulmonary diseases. In this review, we discuss the current evidence on the role of mitochondrial dynamics in pulmonary diseases, with a particular focus on its underlying mechanisms in the development of acute lung injury (ALI)/acute respiratory distress syndrome (ARDS), chronic obstructive pulmonary disease (COPD), asthma, pulmonary fibrosis (PF), pulmonary arterial hypertension (PAH), lung cancer and bronchopulmonary dysplasia (BPD), and outline effective drugs targeting mitochondrial dynamics-related proteins, highlighting the great potential of targeting mitochondrial dynamics in the treatment of pulmonary disease.
Collapse
Affiliation(s)
- Hui Li
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Xinyan Dai
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Junfu Zhou
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Yujuan Wang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Shiying Zhang
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Jiacheng Guo
- Immunotherapy Laboratory, College of Grassland Resources, Southwest Minzu University, Chengdu, Sichuan, China
| | - Lidu Shen
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Hengxiu Yan
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| | - Huiling Jiang
- Immunotherapy Laboratory, College of Pharmacology, Southwest Minzu University, Chengdu, Sichuan, China
| |
Collapse
|
4
|
Godtliebsen G, Larsen KB, Bhujabal Z, Opstad IS, Nager M, Punnakkal AR, Kalstad TB, Olsen R, Lund T, Prasad DK, Agarwal K, Myrmel T, Birgisdottir AB. High-resolution visualization and assessment of basal and OXPHOS-induced mitophagy in H9c2 cardiomyoblasts. Autophagy 2023; 19:2769-2788. [PMID: 37405374 PMCID: PMC10472865 DOI: 10.1080/15548627.2023.2230837] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 06/09/2023] [Accepted: 06/22/2023] [Indexed: 07/06/2023] Open
Abstract
Mitochondria are susceptible to damage resulting from their activity as energy providers. Damaged mitochondria can cause harm to the cell and thus mitochondria are subjected to elaborate quality-control mechanisms including elimination via lysosomal degradation in a process termed mitophagy. Basal mitophagy is a house-keeping mechanism fine-tuning the number of mitochondria according to the metabolic state of the cell. However, the molecular mechanisms underlying basal mitophagy remain largely elusive. In this study, we visualized and assessed the level of mitophagy in H9c2 cardiomyoblasts at basal conditions and after OXPHOS induction by galactose adaptation. We used cells with a stable expression of a pH-sensitive fluorescent mitochondrial reporter and applied state-of-the-art imaging techniques and image analysis. Our data showed a significant increase in acidic mitochondria after galactose adaptation. Using a machine-learning approach we also demonstrated increased mitochondrial fragmentation by OXPHOS induction. Furthermore, super-resolution microscopy of live cells enabled capturing of mitochondrial fragments within lysosomes as well as dynamic transfer of mitochondrial contents to lysosomes. Applying correlative light and electron microscopy we revealed the ultrastructure of the acidic mitochondria confirming their proximity to the mitochondrial network, ER and lysosomes. Finally, exploiting siRNA knockdown strategy combined with flux perturbation with lysosomal inhibitors, we demonstrated the importance of both canonical as well as non-canonical autophagy mediators in lysosomal degradation of mitochondria after OXPHOS induction. Taken together, our high-resolution imaging approaches applied on H9c2 cells provide novel insights on mitophagy during physiologically relevant conditions. The implication of redundant underlying mechanisms highlights the fundamental importance of mitophagy.Abbreviations: ATG: autophagy related; ATG7: autophagy related 7; ATP: adenosine triphosphate; BafA1: bafilomycin A1; CLEM: correlative light and electron microscopy; EGFP: enhanced green fluorescent protein; MAP1LC3B: microtubule associated protein 1 light chain 3 beta; OXPHOS: oxidative phosphorylation; PepA: pepstatin A; PLA: proximity ligation assay; PRKN: parkin RBR E3 ubiquitin protein ligase; RAB5A: RAB5A, member RAS oncogene family; RAB7A: RAB7A, member RAS oncogene family; RAB9A: RAB9A, member RAS oncogene family; ROS: reactive oxygen species; SIM: structured illumination microscopy; siRNA: short interfering RNA; SYNJ2BP: synaptojanin 2 binding protein; TEM: transmission electron microscopy; TOMM20: translocase of outer mitochondrial membrane 20; ULK1: unc-51 like kinase 1.
Collapse
Affiliation(s)
- Gustav Godtliebsen
- Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Kenneth Bowitz Larsen
- Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Department of Medical Biology, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Zambarlal Bhujabal
- Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Ida S. Opstad
- Department of Physics and Technology, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Mireia Nager
- Division of Cardiothoracic and Respiratory Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | | | - Trine B. Kalstad
- Division of Cardiothoracic and Respiratory Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Randi Olsen
- Department of Medical Biology, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Trine Lund
- Department of Medical Biology, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Dilip K. Prasad
- Department of Computer Science, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Krishna Agarwal
- Department of Physics and Technology, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Truls Myrmel
- Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Division of Cardiothoracic and Respiratory Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| | - Asa Birna Birgisdottir
- Department of Clinical Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
- Division of Cardiothoracic and Respiratory Medicine, UiT-The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
5
|
Persad KL, Lopaschuk GD. Energy Metabolism on Mitochondrial Maturation and Its Effects on Cardiomyocyte Cell Fate. Front Cell Dev Biol 2022; 10:886393. [PMID: 35865630 PMCID: PMC9294643 DOI: 10.3389/fcell.2022.886393] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/20/2022] [Indexed: 12/12/2022] Open
Abstract
Alterations in energy metabolism play a major role in the lineage of cardiomyocytes, such as the dramatic changes that occur in the transition from neonate to newborn. As cardiomyocytes mature, they shift from a primarily glycolytic state to a mitochondrial oxidative metabolic state. Metabolic intermediates and metabolites may have epigenetic and transcriptional roles in controlling cell fate by increasing mitochondrial biogenesis. In the maturing cardiomyocyte, such as in the postnatal heart, fatty acid oxidation increases in conjunction with increased mitochondrial biogenesis driven by the transcriptional coregulator PGC1-α. PGC1-α is necessary for mitochondrial biogenesis in the heart at birth, with deficiencies leading to postnatal cardiomyopathy. While stem cell therapy as a treatment for heart failure requires further investigation, studies suggest that adult stem cells may secrete cardioprotective factors which may regulate cardiomyocyte differentiation and survival. This review will discuss how metabolism influences mitochondrial biogenesis and how mitochondrial biogenesis influences cell fate, particularly in the context of the developing cardiomyocyte. The implications of energy metabolism on stem cell differentiation into cardiomyocytes and how this may be utilized as a therapy against heart failure and cardiovascular disease will also be discussed.
Collapse
|
6
|
Liu C, Fu Z, Wu S, Wang X, Zhang S, Chu C, Hong Y, Wu W, Chen S, Jiang Y, Wu Y, Song Y, Liu Y, Guo X. Mitochondrial HSF1 triggers mitochondrial dysfunction and neurodegeneration in Huntington's disease. EMBO Mol Med 2022; 14:e15851. [PMID: 35670111 PMCID: PMC9260212 DOI: 10.15252/emmm.202215851] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 05/10/2022] [Accepted: 05/10/2022] [Indexed: 12/18/2022] Open
Affiliation(s)
- Chunyue Liu
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Zixing Fu
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Shanshan Wu
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Xiaosong Wang
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Shengrong Zhang
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Chu Chu
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Yuan Hong
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Wenbo Wu
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Shengqi Chen
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Yueqing Jiang
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
| | - Yang Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics Key Laboratory of Magnetic Resonance in Biological Systems Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology Chinese Academy of Sciences Wuhan China
| | - Yongbo Song
- Department of Pharmacology Shenyang Pharmaceutical University Shenyang China
| | - Yan Liu
- State Key Laboratory of Reproductive Medicine Interdisciplinary InnoCenter for Organoids Institute for Stem Cell and Neural Regeneration School of Pharmacy Nanjing Medical University Nanjing China
| | - Xing Guo
- State Key Laboratory of Reproductive Medicine Key Laboratory of Human Functional Genomics of Jiangsu Province Department of Neurobiology Interdisciplinary InnoCenter for Organoids School of Basic Medical Sciences Nanjing Medical University Nanjing China
- Department of Endocrinology Sir Run Run Hospital Nanjing Medical University Nanjing Jiangsu China
| |
Collapse
|
7
|
Jin L, Yu B, Liu G, Nie W, Wang J, Chen J, Xiao L, Xia H, Han F, Yang Y. Mitophagy induced by UMI-77 preserves mitochondrial fitness in renal tubular epithelial cells and alleviates renal fibrosis. FASEB J 2022; 36:e22342. [PMID: 35524750 DOI: 10.1096/fj.202200199rr] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/09/2022] [Accepted: 04/25/2022] [Indexed: 12/27/2022]
Abstract
Renal fibrosis is the final common outcome of chronic kidney disease (CKD), which remains a huge challenge due to a lack of targeted treatment. Growing evidence suggests that during the process of CKD, the integrity and function of mitochondria in renal tubular epithelial cells (TECs) are generally impaired and strongly connected with the progression of renal fibrosis. Mitophagy, a selective form of autophagy, could remove aberrant mitochondria to maintain mitochondrial homeostasis. Deficiency of mitophagy has been reported to aggravate renal fibrosis. However, whether induction of mitophagy could alleviate renal fibrosis has not been stated. In this study, we explored the effect of mitophagy activation by UMI-77, a compound recently verified to induce mitophagy, on murine CKD model of unilateral ureteral obstruction (UUO) in vivo and TECs in vitro. In UUO mice, we found the changes of mitochondrial damage, ROS production, transforming growth factor (TGF)-β1/Smad pathway activation, as well as epithelial-mesenchymal transition phenotype and renal fibrosis, and these changes were ameliorated by mitophagy enhancement using UMI-77. Moreover, TEC apoptosis, nuclear factor (NF)-κB signaling activation, and interstitial inflammation after UUO were significantly mitigated by augmented mitophagy. Then, we found UMI-77 could effectively and safely induce mitophagy in TECs in vitro, and reduced TGF-β1/Smad signaling and downstream profibrotic responses in TGF-β1-treated TECs. These changes were restored by a mitophagy inhibitor. In conclusion, we demonstrated that mitophagy activation protected against renal fibrosis through improving mitochondrial fitness, downregulating TGF-β1/Smad signaling and alleviating TEC injuries and inflammatory infiltration in kidneys.
Collapse
Affiliation(s)
- Lini Jin
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Binfeng Yu
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Guangjun Liu
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Wanyun Nie
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Junni Wang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Jianghua Chen
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Liang Xiao
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Hongguang Xia
- Liangzhu Laboratory, Zhejiang University Medical Center, Department of Biochemistry & Research Center of Clinical Pharmacy of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Fei Han
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| | - Yi Yang
- Key Laboratory of Kidney Disease Prevention and Control Technology, Kidney Disease Center, Institute of Nephrology, the First Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
8
|
Bordoni M, Pansarasa O, Scarian E, Cristofani R, Leone R, Fantini V, Garofalo M, Diamanti L, Bernuzzi S, Gagliardi S, Carelli S, Poletti A, Cereda C. Lysosomes Dysfunction Causes Mitophagy Impairment in PBMCs of Sporadic ALS Patients. Cells 2022; 11:cells11081272. [PMID: 35455952 PMCID: PMC9030813 DOI: 10.3390/cells11081272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 04/06/2022] [Accepted: 04/07/2022] [Indexed: 02/06/2023] Open
Abstract
Mitochondria alterations are present in tissues derived from patients and animal models, but no data are available for peripheral blood mononuclear cells (PBMCs) of ALS patients. This work aims to investigate mitophagy in PBMCs of sporadic (sALS) patients and how this pathway can be tuned by using small molecules. We found the presence of morphologically atypical mitochondria by TEM and morphological abnormalities by MitoTracker™. We found a decreased number of healthy mitochondria in sALS PBMCs and an impairment of mitophagy with western blot and immunofluorescence. After rapamycin treatment, we found a higher increase in the LC3 marker in sALS PBMCs, while after NH4Cl treatment, we found a lower increase in the LC3 marker. Finally, mTOR-independent autophagy induction with trehalose resulted in a significant decrease in the lysosomes level sALS PBMCs. Our data suggest that the presence of morphologically altered mitochondria and an inefficient turnover of damaged mitochondria in PBMCs of sALS patients rely on the impairment of the mitophagy pathway. We also found that the induction of the mTOR-independent autophagy pathway leads to a decrease in lysosomes level, suggesting a more sensitivity of sALS PBMCs to trehalose. Such evidence suggests that trehalose could represent an effective treatment for ALS patients.
Collapse
Affiliation(s)
- Matteo Bordoni
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Orietta Pansarasa
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
- Correspondence: ; Tel.: +0382-380-248
| | - Eveljn Scarian
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy
| | - Riccardo Cristofani
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Università Degli Studi di Milano, 20133 Milano, Italy; (R.C.); (A.P.)
| | | | - Valentina Fantini
- Laboratory of Neurobiology and Neurogenetic, Golgi-Cenci Foundation, 20081 Abbiategrasso, Italy;
| | - Maria Garofalo
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Luca Diamanti
- Neuroncology Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy;
| | - Stefano Bernuzzi
- Immunohematological and Transfusional Service and Centre of Transplantation Immunology, IRCCS “San Matteo Foundation”, 27100 Pavia, Italy;
| | - Stella Gagliardi
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| | - Stephana Carelli
- Department of Biomedical and Clinical Sciences “L. Sacco”, University of Milan, 20157 Milan, Italy;
- Pediatric Clinical Research Centre Fondazione “Romeo ed Enrica Invernizzi”, University of Milano, 20157 Milan, Italy
| | - Angelo Poletti
- Dipartimento di Scienze Farmacologiche e Biomolecolari (DiSFeB), Dipartimento di Eccellenza 2018-2022, Università Degli Studi di Milano, 20133 Milano, Italy; (R.C.); (A.P.)
| | - Cristina Cereda
- Genomic and Post-Genomic Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy; (M.B.); (E.S.); (M.G.); (S.G.); (C.C.)
| |
Collapse
|
9
|
Heidorn-Czarna M, Maziak A, Janska H. Protein Processing in Plant Mitochondria Compared to Yeast and Mammals. FRONTIERS IN PLANT SCIENCE 2022; 13:824080. [PMID: 35185991 PMCID: PMC8847149 DOI: 10.3389/fpls.2022.824080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 01/12/2022] [Indexed: 05/02/2023]
Abstract
Limited proteolysis, called protein processing, is an essential post-translational mechanism that controls protein localization, activity, and in consequence, function. This process is prevalent for mitochondrial proteins, mainly synthesized as precursor proteins with N-terminal sequences (presequences) that act as targeting signals and are removed upon import into the organelle. Mitochondria have a distinct and highly conserved proteolytic system that includes proteases with sole function in presequence processing and proteases, which show diverse mitochondrial functions with limited proteolysis as an additional one. In virtually all mitochondria, the primary processing of N-terminal signals is catalyzed by the well-characterized mitochondrial processing peptidase (MPP). Subsequently, a second proteolytic cleavage occurs, leading to more stabilized residues at the newly formed N-terminus. Lately, mitochondrial proteases, intermediate cleavage peptidase 55 (ICP55) and octapeptidyl protease 1 (OCT1), involved in proteolytic cleavage after MPP and their substrates have been described in the plant, yeast, and mammalian mitochondria. Mitochondrial proteins can also be processed by removing a peptide from their N- or C-terminus as a maturation step during insertion into the membrane or as a regulatory mechanism in maintaining their function. This type of limited proteolysis is characteristic for processing proteases, such as IMP and rhomboid proteases, or the general mitochondrial quality control proteases ATP23, m-AAA, i-AAA, and OMA1. Identification of processing protease substrates and defining their consensus cleavage motifs is now possible with the help of large-scale quantitative mass spectrometry-based N-terminomics, such as combined fractional diagonal chromatography (COFRADIC), charge-based fractional diagonal chromatography (ChaFRADIC), or terminal amine isotopic labeling of substrates (TAILS). This review summarizes the current knowledge on the characterization of mitochondrial processing peptidases and selected N-terminomics techniques used to uncover protease substrates in the plant, yeast, and mammalian mitochondria.
Collapse
|
10
|
Luan Y, Ren KD, Luan Y, Chen X, Yang Y. Mitochondrial Dynamics: Pathogenesis and Therapeutic Targets of Vascular Diseases. Front Cardiovasc Med 2021; 8:770574. [PMID: 34938787 PMCID: PMC8685340 DOI: 10.3389/fcvm.2021.770574] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2021] [Accepted: 10/25/2021] [Indexed: 12/12/2022] Open
Abstract
Vascular diseases, particularly atherosclerosis, are associated with high morbidity and mortality. Endothelial cell (EC) or vascular smooth muscle cell (VSMC) dysfunction leads to blood vessel abnormalities, which cause a series of vascular diseases. The mitochondria are the core sites of cell energy metabolism and function in blood vessel development and vascular disease pathogenesis. Mitochondrial dynamics, including fusion and fission, affect a variety of physiological or pathological processes. Multiple studies have confirmed the influence of mitochondrial dynamics on vascular diseases. This review discusses the regulatory mechanisms of mitochondrial dynamics, the key proteins that mediate mitochondrial fusion and fission, and their potential effects on ECs and VSMCs. We demonstrated the possibility of mitochondrial dynamics as a potential target for the treatment of vascular diseases.
Collapse
Affiliation(s)
- Yi Luan
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Kai-Di Ren
- Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Henan Key Laboratory of Precision Clinical Pharmacy, Zhengzhou University, Zhengzhou, China
| | - Ying Luan
- Department of Physiology and Neurobiology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xing Chen
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Yang
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
11
|
Yang X, Ha G, Needleman D. A coarse-grained NADH redox model enables inference of subcellular metabolic fluxes from fluorescence lifetime imaging. eLife 2021; 10:73808. [PMID: 34806591 PMCID: PMC8935353 DOI: 10.7554/elife.73808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/19/2021] [Indexed: 11/13/2022] Open
Abstract
Mitochondrial metabolism is of central importance to diverse aspects of cell and developmental biology. Defects in mitochondria are associated with many diseases, including cancer, neuropathology, and infertility. Our understanding of mitochondrial metabolism in situ and dysfunction in diseases are limited by the lack of techniques to measure mitochondrial metabolic fluxes with sufficient spatiotemporal resolution. Herein, we developed a new method to infer mitochondrial metabolic fluxes in living cells with subcellular resolution from fluorescence lifetime imaging of NADH. This result is based on the use of a generic coarse-grained NADH redox model. We tested the model in mouse oocytes and human tissue culture cells subject to a wide variety of perturbations by comparing predicted fluxes through the electron transport chain (ETC) to direct measurements of oxygen consumption rate. Interpreting the fluorescence lifetime imaging microscopy measurements of NADH using this model, we discovered a homeostasis of ETC flux in mouse oocytes: perturbations of nutrient supply and energy demand of the cell do not change ETC flux despite significantly impacting NADH metabolic state. Furthermore, we observed a subcellular spatial gradient of ETC flux in mouse oocytes and found that this gradient is primarily a result of a spatially heterogeneous mitochondrial proton leak. We concluded from these observations that ETC flux in mouse oocytes is not controlled by energy demand or supply, but by the intrinsic rates of mitochondrial respiration.
Collapse
Affiliation(s)
- Xingbo Yang
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Gloria Ha
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| | - Dan Needleman
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, United States
| |
Collapse
|
12
|
Liu YT, Huang X, Nguyen D, Shammas MK, Wu BP, Dombi E, Springer DA, Poulton J, Sekine S, Narendra DP. Loss of CHCHD2 and CHCHD10 activates OMA1 peptidase to disrupt mitochondrial cristae phenocopying patient mutations. Hum Mol Genet 2021; 29:1547-1567. [PMID: 32338760 DOI: 10.1093/hmg/ddaa077] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 03/24/2020] [Accepted: 04/20/2020] [Indexed: 11/13/2022] Open
Abstract
Dominant mutations in the mitochondrial paralogs coiled-helix-coiled-helix (CHCHD) domain 2 (C2) and CHCHD10 (C10) were recently identified as causing Parkinson's disease and amyotrophic lateral sclerosis/frontotemporal dementia/myopathy, respectively. The mechanism by which they disrupt mitochondrial cristae, however, has been uncertain. Using the first C2/C10 double knockout (DKO) mice, we report that C10 pathogenesis and the normal function of C2/C10 are intimately linked. Similar to patients with C10 mutations, we found that C2/C10 DKO mice have disrupted mitochondrial cristae, because of cleavage of the mitochondrial-shaping protein long form of OPA1 (L-OPA1) by the stress-induced peptidase OMA1. OMA1 was found to be activated similarly in affected tissues of mutant C10 knock-in (KI) mice, demonstrating that L-OPA1 cleavage is a novel mechanism for cristae abnormalities because of both C10 mutation and C2/C10 loss. Using OMA1 activation as a functional assay, we found that C2 and C10 are partially functionally redundant, and some but not all disease-causing mutations have retained activity. Finally, C2/C10 DKO mice partially phenocopied mutant C10 KI mice with the development of cardiomyopathy and activation of the integrated mitochondrial integrated stress response in affected tissues, tying mutant C10 pathogenesis to C2/C10 function.
Collapse
Affiliation(s)
- Yi-Ting Liu
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Xiaoping Huang
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Diana Nguyen
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Mario K Shammas
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Beverly P Wu
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eszter Dombi
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| | - Danielle A Springer
- Murine Phenotyping Core, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Joanna Poulton
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford OX3 9DU, UK
| | - Shiori Sekine
- Aging Institute, Division of Cardiology, Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Derek P Narendra
- Inherited Movement Disorders Unit, Neurogenetics Branch, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
13
|
Abudu YP, Shrestha BK, Zhang W, Palara A, Brenne HB, Larsen KB, Wolfson DL, Dumitriu G, Øie CI, Ahluwalia BS, Levy G, Behrends C, Tooze SA, Mouilleron S, Lamark T, Johansen T. SAMM50 acts with p62 in piecemeal basal- and OXPHOS-induced mitophagy of SAM and MICOS components. J Cell Biol 2021; 220:212185. [PMID: 34037656 PMCID: PMC8160579 DOI: 10.1083/jcb.202009092] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 03/24/2021] [Accepted: 04/28/2021] [Indexed: 12/21/2022] Open
Abstract
Mitophagy is the degradation of surplus or damaged mitochondria by autophagy. In addition to programmed and stress-induced mitophagy, basal mitophagy processes exert organelle quality control. Here, we show that the sorting and assembly machinery (SAM) complex protein SAMM50 interacts directly with ATG8 family proteins and p62/SQSTM1 to act as a receptor for a basal mitophagy of components of the SAM and mitochondrial contact site and cristae organizing system (MICOS) complexes. SAMM50 regulates mitochondrial architecture by controlling formation and assembly of the MICOS complex decisive for normal cristae morphology and exerts quality control of MICOS components. To this end, SAMM50 recruits ATG8 family proteins through a canonical LIR motif and interacts with p62/SQSTM1 to mediate basal mitophagy of SAM and MICOS components. Upon metabolic switch to oxidative phosphorylation, SAMM50 and p62 cooperate to mediate efficient mitophagy.
Collapse
Affiliation(s)
- Yakubu Princely Abudu
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Birendra Kumar Shrestha
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Wenxin Zhang
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London, UK
| | - Anthimi Palara
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Hanne Britt Brenne
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Kenneth Bowitz Larsen
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Deanna Lynn Wolfson
- Department of Physics and Technology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Gianina Dumitriu
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Cristina Ionica Øie
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Balpreet Singh Ahluwalia
- Department of Physics and Technology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Gahl Levy
- Vascular Biology Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Christian Behrends
- Institute of Biochemistry II, Goethe University Hospital, Frankfurt am Main, Germany.,Munich Cluster for Systems Neurology (SyNergy), Ludwig Maximilian University, Munich, Germany
| | - Sharon A Tooze
- Molecular Cell Biology of Autophagy Laboratory, The Francis Crick Institute, London, UK
| | - Stephane Mouilleron
- Structural Biology Science Technology Platform, The Francis Crick Institute, London, UK
| | - Trond Lamark
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| | - Terje Johansen
- Molecular Cancer Research Group, Department of Medical Biology, University of Tromsø-The Arctic University of Norway, Tromsø, Norway
| |
Collapse
|
14
|
Changes in Drp1 Function and Mitochondrial Morphology Are Associated with the α-Synuclein Pathology in a Transgenic Mouse Model of Parkinson's Disease. Cells 2021; 10:cells10040885. [PMID: 33924585 PMCID: PMC8070398 DOI: 10.3390/cells10040885] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 04/07/2021] [Accepted: 04/08/2021] [Indexed: 12/16/2022] Open
Abstract
Alterations in mitochondrial function and morphology are associated with many human diseases, including cancer and neurodegenerative diseases. Mitochondrial impairment is linked to Parkinson's disease (PD) pathogenesis, and alterations in mitochondrial dynamics are seen in PD models. In particular, α-synuclein (αS) abnormalities are often associated with pathological changes to mitochondria. However, the relationship between αS pathology and mitochondrial dynamics remains poorly defined. Herein, we examined a mouse model of α-synucleinopathy for αS pathology-linked alterations in mitochondrial dynamics in vivo. We show that α-synucleinopathy in a transgenic (Tg) mouse model expressing familial PD-linked mutant A53T human αS (TgA53T) is associated with a decrease in Drp1 localization and activity in the mitochondria. In addition, we show that the loss of Drp1 function in the mitochondria is associated with two distinct phenotypes of enlarged neuronal mitochondria. Mitochondrial enlargement was only present in diseased animals and, apart from Drp1, other proteins involved in mitochondrial dynamics are unlikely to cause these changes, as their levels remained mostly unchanged. Further, the levels of Mfn1, a protein that facilitates mitochondrial fusion, was decreased nonspecifically with transgene expression. These results support the view that altered mitochondrial dynamics are a significant neuropathological factor in α-synucleinopathies.
Collapse
|
15
|
Hogarth K, Vanama RB, Stratmann G, Maynes JT. Singular and short-term anesthesia exposure in the developing brain induces persistent neuronal changes consistent with chronic neurodegenerative disease. Sci Rep 2021; 11:5673. [PMID: 33707598 PMCID: PMC7952562 DOI: 10.1038/s41598-021-85125-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 02/25/2021] [Indexed: 01/31/2023] Open
Abstract
The potential adverse impact of inhalational anesthetics on the developing brain was highlighted by the addition of a medication warning by the U.S. Food and Drug Administration for their use in the pediatric population. To investigate mechanisms by which early life anesthesia exposure could induce long-term neuronal dysfunction, we exposed rats to 1 minimum alveolar concentration sevoflurane at 7 days of life. The animals were raised normally until adulthood (P300) prior to sacrifice and analysis of cortical tissue structure (TEM), mitochondrial quality control and biogenesis pathways (Western blot, ELISA, ADP/ATP content), and markers of oxidative stress, proteotoxicity and inflammation (Western blot, ELISA). We found that early life anesthesia exposure led to adverse changes in mitochondrial quality maintenance pathways, autophagy and mitochondrial biogenesis. Although there was an escalation of oxidative stress markers and an increase in the nuclear localization of stress-related transcription factors, cellular redox compensatory responses were blunted, and oxidative phosphorylation was reduced. We found upregulation of mitochondrial stress and proteotoxicity markers, but a significant reduction of mitochondrial unfolded protein response end-effectors, contributing to an increase in inflammation. Contrary to acute exposure, we did not find an increase in apoptosis. Our findings suggest that a limited, early exposure to anesthesia may produce lasting cellular dysfunction through the induction of a sustained energy deficient state, resulting in persistent neuroinflammation and altered proteostasis/toxicity, mimicking aspects of chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- Kaley Hogarth
- Division of Molecular Medicine, SickKids Research Institute, Toronto, Canada
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, 555 University Ave., Toronto, ON, M5G 1X8, Canada
| | - Ramesh Babu Vanama
- Division of Molecular Medicine, SickKids Research Institute, Toronto, Canada
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, 555 University Ave., Toronto, ON, M5G 1X8, Canada
| | - Greg Stratmann
- Department of Anesthesia and Perioperative Medicine, University of California, San Francisco, San Francisco, USA
| | - Jason T Maynes
- Division of Molecular Medicine, SickKids Research Institute, Toronto, Canada.
- Department of Anesthesia and Pain Medicine, Hospital for Sick Children, 555 University Ave., Toronto, ON, M5G 1X8, Canada.
- Department of Anesthesiology and Pain Medicine, University of Toronto, Toronto, Canada.
| |
Collapse
|
16
|
Garcia I, Calderon F, la Torre PD, Vallier SS, Rodriguez C, Agarwala D, Keniry M, Innis-Whitehouse W, Gilkerson R. Mitochondrial OPA1 cleavage is reversibly activated by differentiation of H9c2 cardiomyoblasts. Mitochondrion 2021; 57:88-96. [PMID: 33383158 PMCID: PMC7904612 DOI: 10.1016/j.mito.2020.12.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 12/17/2020] [Accepted: 12/21/2020] [Indexed: 11/17/2022]
Abstract
Optic atrophy-1 (OPA1) is a dynamin-like GTPase localized to the mitochondrial inner membrane, playing key roles in inner membrane fusion and cristae maintenance. OPA1 is regulated by the mitochondrial transmembrane potential (Δψm): when Δψm is intact, long OPA1 isoforms (L-OPA1) carry out inner membrane fusion. Upon loss of Δψm, L-OPA1 isoforms are proteolytically cleaved to short (S-OPA1) isoforms by the stress-inducible OMA1 metalloprotease, causing collapse of the mitochondrial network and promoting apoptosis. Here, we show that L-OPA1 isoforms of H9c2 cardiomyoblasts are retained under loss of Δψm, despite the presence of OMA1. However, when H9c2s are differentiated to a more cardiac-like phenotype via treatment with retinoic acid (RA) in low serum media, loss of Δ ψm induces robust, and reversible, cleavage of L-OPA1 and subsequent OMA1 degradation. These findings indicate that a potent developmental switch regulates Δ ψm-sensitive OPA1 cleavage, suggesting novel developmental and regulatory mechanisms for OPA1 homeostasis.
Collapse
Affiliation(s)
- Iraselia Garcia
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA; Department of Biology, South Texas College, McAllen, TX, USA
| | - Fredy Calderon
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Patrick De la Torre
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Shaynah St Vallier
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Cristobal Rodriguez
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Divya Agarwala
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | - Megan Keniry
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA
| | | | - Robert Gilkerson
- Department of Biology, The University of Texas Rio Grande Valley, Edinburg, TX, USA; Clinical Laboratory Sciences, The University of Texas Rio Grande Valley, Edinburg, TX, USA.
| |
Collapse
|
17
|
Douida A, Batista F, Boto P, Regdon Z, Robaszkiewicz A, Tar K. Cells Lacking PA200 Adapt to Mitochondrial Dysfunction by Enhancing Glycolysis via Distinct Opa1 Processing. Int J Mol Sci 2021; 22:ijms22041629. [PMID: 33562813 PMCID: PMC7914502 DOI: 10.3390/ijms22041629] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 02/01/2021] [Accepted: 02/02/2021] [Indexed: 02/07/2023] Open
Abstract
The conserved Blm10/PA200 proteins are proteasome activators. Previously, we identified PA200-enriched regions in the genome of SH-SY5Y neuroblastoma cells by chromatin immunoprecipitation (ChIP) and ChIP-seq analysis. We also found that selective mitochondrial inhibitors induced PA200 redistribution in the genome. Collectively, our data indicated that PA200 regulates cellular homeostasis at the transcriptional level. In the present study, our aim is to investigate the impact of stable PA200 depletion (shPA200) on the overall transcriptome of SH-SY5Y cells. RNA-seq data analysis reveals that the genetic ablation of PA200 leads to overall changes in the transcriptional landscape of SH-SY5Y neuroblastoma cells. PA200 activates and represses genes regulating metabolic processes, such as the glycolysis and mitochondrial function. Using metabolic assays in live cells, we showed that stable knockdown of PA200 does not change basal respiration. Spare respiratory capacity and proton leak however are slightly, yet significantly, reduced in PA200-deficient cells by 99.834% and 84.147%, respectively, compared to control. Glycolysis and glycolytic capacity show a 42.186% and 26.104% increase in shPA200 cells, respectively, compared to control. These data suggest a shift from oxidative phosphorylation to glycolysis especially when cells are exposed to oligomycin-induced stress. Furthermore, we observed a preserved long and compact tubular mitochondrial morphology after inhibition of ATP synthase by oligomycin, which might be associated with the glycolytic change of shPA200 cells. The present study also demonstrates that the proteolytic cleavage of Opa1 is affected, and that the level of OMA1 is significantly reduced in shPA200 cells upon oligomycin-induced mitochondrial insult. Together, these findings suggest a role for PA200 in the regulation of metabolic changes in response to selective inhibition of ATP synthase in an in vitro cellular model.
Collapse
Affiliation(s)
- Abdennour Douida
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (A.D.); (Z.R.)
- Doctoral School of Molecular Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Frank Batista
- Department of Biochemistry and Molecular Biology, Faculty of Medicine, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Pal Boto
- Stem Cell Differentiation Laboratory, Department of Biochemistry and Molecular Biology, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Zsolt Regdon
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (A.D.); (Z.R.)
- Doctoral School of Molecular Medicine, University of Debrecen, H-4032 Debrecen, Hungary
| | - Agnieszka Robaszkiewicz
- Department of General Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, 90-236 Lodz, Poland;
| | - Krisztina Tar
- Department of Medical Chemistry, Faculty of Medicine, University of Debrecen, Egyetem tér 1, H-4032 Debrecen, Hungary; (A.D.); (Z.R.)
- Correspondence: ; Tel.: +36-52-412-345
| |
Collapse
|
18
|
Cioffi F, Adam RHI, Broersen K. Molecular Mechanisms and Genetics of Oxidative Stress in Alzheimer's Disease. J Alzheimers Dis 2020; 72:981-1017. [PMID: 31744008 PMCID: PMC6971833 DOI: 10.3233/jad-190863] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Alzheimer’s disease is the most common neurodegenerative disorder that can cause dementia in elderly over 60 years of age. One of the disease hallmarks is oxidative stress which interconnects with other processes such as amyloid-β deposition, tau hyperphosphorylation, and tangle formation. This review discusses current thoughts on molecular mechanisms that may relate oxidative stress to Alzheimer’s disease and identifies genetic factors observed from in vitro, in vivo, and clinical studies that may be associated with Alzheimer’s disease-related oxidative stress.
Collapse
Affiliation(s)
- Federica Cioffi
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Rayan Hassan Ibrahim Adam
- Nanobiophysics Group, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| | - Kerensa Broersen
- Applied Stem Cell Technologies, Technical Medical Centre, Faculty of Science and Technology, University of Twente, Enschede, The Netherlands
| |
Collapse
|
19
|
Lechuga-Vieco AV, Latorre-Pellicer A, Johnston IG, Prota G, Gileadi U, Justo-Méndez R, Acín-Pérez R, Martínez-de-Mena R, Fernández-Toro JM, Jimenez-Blasco D, Mora A, Nicolás-Ávila JA, Santiago DJ, Priori SG, Bolaños JP, Sabio G, Criado LM, Ruíz-Cabello J, Cerundolo V, Jones NS, Enríquez JA. Cell identity and nucleo-mitochondrial genetic context modulate OXPHOS performance and determine somatic heteroplasmy dynamics. SCIENCE ADVANCES 2020; 6:eaba5345. [PMID: 32832682 PMCID: PMC7439646 DOI: 10.1126/sciadv.aba5345] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 06/17/2020] [Indexed: 05/02/2023]
Abstract
Heteroplasmy, multiple variants of mitochondrial DNA (mtDNA) in the same cytoplasm, may be naturally generated by mutations but is counteracted by a genetic mtDNA bottleneck during oocyte development. Engineered heteroplasmic mice with nonpathological mtDNA variants reveal a nonrandom tissue-specific mtDNA segregation pattern, with few tissues that do not show segregation. The driving force for this dynamic complex pattern has remained unexplained for decades, challenging our understanding of this fundamental biological problem and hindering clinical planning for inherited diseases. Here, we demonstrate that the nonrandom mtDNA segregation is an intracellular process based on organelle selection. This cell type-specific decision arises jointly from the impact of mtDNA haplotypes on the oxidative phosphorylation (OXPHOS) system and the cell metabolic requirements and is strongly sensitive to the nuclear context and to environmental cues.
Collapse
Affiliation(s)
- Ana Victoria Lechuga-Vieco
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- CIBERES: C/ Melchor Fernández-Almagro 3, 28029 Madrid, Spain
| | - Ana Latorre-Pellicer
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Unit of Clinical Genetics and Functional Genomics, Department of Pharmacology-Physiology, School of Medicine, University of Zaragoza, IIS Aragon, E-50009 Zaragoza, Spain
| | - Iain G. Johnston
- Department of Mathematics, Faculty of Mathematics and Natural Sciences, University of Bergen, Bergen, Norway
| | - Gennaro Prota
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Uzi Gileadi
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Raquel Justo-Méndez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Rebeca Acín-Pérez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | | | | | - Daniel Jimenez-Blasco
- IBFG, Universidad de Salamanca, Salamanca, Spain
- IBSAL, Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, Salamanca, Spain
- CIBERFES, C/Melchor Fernández-Almagro 3, 28029 Madrid, Spain
| | - Alfonso Mora
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | | | - Demetrio J. Santiago
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Molecular Cardiology, IRCCS ICS Maugeri, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Silvia G. Priori
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- Molecular Cardiology, IRCCS ICS Maugeri, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Juan Pedro Bolaños
- IBFG, Universidad de Salamanca, Salamanca, Spain
- IBSAL, Hospital Universitario de Salamanca, Universidad de Salamanca, CSIC, Salamanca, Spain
- CIBERFES, C/Melchor Fernández-Almagro 3, 28029 Madrid, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Luis Miguel Criado
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
| | - Jesús Ruíz-Cabello
- CIBERES: C/ Melchor Fernández-Almagro 3, 28029 Madrid, Spain
- CIC biomaGUNE 20014 Donostia/San Sebastián, Gipuzkoa, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Universidad Complutense Madrid, Madrid, Spain
| | - Vincenzo Cerundolo
- MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Nick S. Jones
- EPSRC Centre for the Mathematics of Precision Healthcare, Department of Mathematics, Imperial College London, London SW7 2BB, UK
| | - José Antonio Enríquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, Madrid, Spain
- CIBERFES, C/Melchor Fernández-Almagro 3, 28029 Madrid, Spain
| |
Collapse
|
20
|
Ge P, Dawson VL, Dawson TM. PINK1 and Parkin mitochondrial quality control: a source of regional vulnerability in Parkinson's disease. Mol Neurodegener 2020; 15:20. [PMID: 32169097 PMCID: PMC7071653 DOI: 10.1186/s13024-020-00367-7] [Citation(s) in RCA: 263] [Impact Index Per Article: 65.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 02/13/2020] [Indexed: 02/06/2023] Open
Abstract
That certain cell types in the central nervous system are more likely to undergo neurodegeneration in Parkinson's disease is a widely appreciated but poorly understood phenomenon. Many vulnerable subpopulations, including dopamine neurons in the substantia nigra pars compacta, have a shared phenotype of large, widely distributed axonal networks, dense synaptic connections, and high basal levels of neural activity. These features come at substantial bioenergetic cost, suggesting that these neurons experience a high degree of mitochondrial stress. In such a context, mechanisms of mitochondrial quality control play an especially important role in maintaining neuronal survival. In this review, we focus on understanding the unique challenges faced by the mitochondria in neurons vulnerable to neurodegeneration in Parkinson's and summarize evidence that mitochondrial dysfunction contributes to disease pathogenesis and to cell death in these subpopulations. We then review mechanisms of mitochondrial quality control mediated by activation of PINK1 and Parkin, two genes that carry mutations associated with autosomal recessive Parkinson's disease. We conclude by pinpointing critical gaps in our knowledge of PINK1 and Parkin function, and propose that understanding the connection between the mechanisms of sporadic Parkinson's and defects in mitochondrial quality control will lead us to greater insights into the question of selective vulnerability.
Collapse
Affiliation(s)
- Preston Ge
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Department of Neurology, Department of Physiology, Solomon H. Snyder Department of Neuroscience, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 733 North Broadway, Suite 731, Baltimore, MD 21205 USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130 USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130 USA
- Present address: Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139 USA
- Present address: Picower Institute for Learning and Memory, Cambridge, MA 02139 USA
- Present address: Harvard-MIT MD/PhD Program, Harvard Medical School, Boston, MA 02115 USA
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Department of Neurology, Department of Physiology, Solomon H. Snyder Department of Neuroscience, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 733 North Broadway, Suite 731, Baltimore, MD 21205 USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130 USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130 USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Department of Neurology, Department of Physiology, Solomon H. Snyder Department of Neuroscience, Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, 733 North Broadway, Suite 731, Baltimore, MD 21205 USA
- Adrienne Helis Malvin Medical Research Foundation, New Orleans, LA 70130 USA
- Diana Helis Henry Medical Research Foundation, New Orleans, LA 70130 USA
| |
Collapse
|
21
|
Zhao JF, Rodger CE, Allen GFG, Weidlich S, Ganley IG. HIF1α-dependent mitophagy facilitates cardiomyoblast differentiation. Cell Stress 2020; 4:99-113. [PMID: 32420530 PMCID: PMC7212530 DOI: 10.15698/cst2020.05.220] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 02/20/2020] [Accepted: 02/25/2020] [Indexed: 01/12/2023] Open
Abstract
Mitophagy is thought to play a key role in eliminating damaged mitochondria, with diseases such as cancer and neurodegeneration exhibiting defects in this process. Mitophagy is also involved in cell differentiation and maturation, potentially through modulating mitochondrial metabolic reprogramming. Here we examined mitophagy that is induced upon iron chelation and found that the transcriptional activity of HIF1α, in part through upregulation of BNIP3 and NIX, is an essential mediator of this pathway in SH-SY5Y cells. In contrast, HIF1α is dispensable for mitophagy occurring upon mitochondrial depolarisation. To examine the role of this pathway in a metabolic reprogramming and differentiation context, we utilised the H9c2 cell line model of cardiomyocyte maturation. During differentiation of these cardiomyoblasts, mitophagy increased and required HIF1α-dependent upregulation of NIX. Though HIF1α was essential for expression of key cardiomyocyte markers, mitophagy was not directly required. However, enhancing mitophagy through NIX overexpression, accelerated marker gene expression. Taken together, our findings provide a molecular link between mitophagy signalling and cardiomyocyte differentiation and suggest that although mitophagy may not be essential per se, it plays a critical role in maintaining mitochondrial integrity during this energy demanding process.
Collapse
Affiliation(s)
- Jin-Feng Zhao
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
- Equal contribution
| | - Catherine E Rodger
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
- Equal contribution
| | - George F G Allen
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Simone Weidlich
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| | - Ian G Ganley
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, DD1 5EH, UK
| |
Collapse
|
22
|
Galkina KV, Zyrina AN, Golyshev SA, Kashko ND, Markova OV, Sokolov SS, Severin FF, Knorre DA. Mitochondrial dynamics in yeast with repressed adenine nucleotide translocator AAC2. Eur J Cell Biol 2020; 99:151071. [PMID: 32057484 DOI: 10.1016/j.ejcb.2020.151071] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 01/30/2020] [Accepted: 01/30/2020] [Indexed: 12/25/2022] Open
Abstract
The mitochondrial network structure dynamically adapts to cellular metabolic challenges. Mitochondrial depolarisation, particularly, induces fragmentation of the network. This fragmentation may be a result of either a direct regulation of the mitochondrial fusion machinery by transmembrane potential or an indirect effect of metabolic remodelling. Activities of ATP synthase and adenine nucleotide translocator (ANT) link the mitochondrial transmembrane potential with the cytosolic NTP/NDP ratio. Given that mitochondrial fusion requires cytosolic GTP, a decrease in the NTP/NDP ratio might also account for protonophore-induced mitochondrial fragmentation. For evaluating the contributions of direct and indirect mechanisms to mitochondrial remodelling, we assessed the morphology of the mitochondrial network in yeast cells with inhibited ANT. We showed that the repression of AAC2 (PET9), a major ANT gene in yeast, increases mitochondrial transmembrane potential. However, the mitochondrial network in this strain was fragmented. Meanwhile, AAC2 repression did not prevent mitochondrial fusion in zygotes; nor did it inhibit mitochondrial hyperfusion induced by Dnm1p inhibitor mdivi-1. These results suggest that the inhibition of ANT, rather than preventing mitochondrial fusion, facilitates mitochondrial fission. The protonophores were not able to induce additional mitochondrial fragmentation in an AAC2-repressed strain and in yeast cells with inhibited ATP synthase. Importantly, treatment with the ATP synthase inhibitor oligomycin A also induced mitochondrial fragmentation and hyperpolarization. Taken together, our data suggest that ATP/ADP translocation plays a crucial role in shaping of the mitochondrial network and exemplify that an increase in mitochondrial membrane potential does not necessarily oppose mitochondrial fragmentation.
Collapse
Affiliation(s)
- Kseniia V Galkina
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskiye Gory 1-73, Moscow, 119991, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskiye Gory 1-40, Moscow, 119991, Russia
| | - Anna N Zyrina
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskiye Gory 1-40, Moscow, 119991, Russia
| | - Sergey A Golyshev
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskiye Gory 1-40, Moscow, 119991, Russia
| | - Nataliia D Kashko
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Leninskiye Gory 1-73, Moscow, 119991, Russia
| | - Olga V Markova
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskiye Gory 1-40, Moscow, 119991, Russia
| | - Svyatoslav S Sokolov
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskiye Gory 1-40, Moscow, 119991, Russia
| | - Fedor F Severin
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskiye Gory 1-40, Moscow, 119991, Russia
| | - Dmitry A Knorre
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Leninskiye Gory 1-40, Moscow, 119991, Russia; Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, 119991, Russia.
| |
Collapse
|
23
|
Yu R, Lendahl U, Nistér M, Zhao J. Regulation of Mammalian Mitochondrial Dynamics: Opportunities and Challenges. Front Endocrinol (Lausanne) 2020; 11:374. [PMID: 32595603 PMCID: PMC7300174 DOI: 10.3389/fendo.2020.00374] [Citation(s) in RCA: 98] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/12/2020] [Indexed: 01/01/2023] Open
Abstract
Mitochondria are highly dynamic organelles and important for a variety of cellular functions. They constantly undergo fission and fusion events, referred to as mitochondrial dynamics, which affects the shape, size, and number of mitochondria in the cell, as well as mitochondrial subcellular transport, mitochondrial quality control (mitophagy), and programmed cell death (apoptosis). Dysfunctional mitochondrial dynamics is associated with various human diseases. Mitochondrial dynamics is mediated by a set of mitochondria-shaping proteins in both yeast and mammals. In this review, we describe recent insights into the potential molecular mechanisms underlying mitochondrial fusion and fission, particularly highlighting the coordinating roles of different mitochondria-shaping proteins in the processes, as well as the roles of the endoplasmic reticulum (ER), the actin cytoskeleton and membrane phospholipids in the regulation of mitochondrial dynamics. We particularly focus on emerging roles for the mammalian mitochondrial proteins Fis1, Mff, and MIEFs (MIEF1 and MIEF2) in regulating the recruitment of the cytosolic Drp1 to the surface of mitochondria and how these proteins, especially Fis1, mediate crosstalk between the mitochondrial fission and fusion machineries. In summary, this review provides novel insights into the molecular mechanisms of mammalian mitochondrial dynamics and the involvement of these mechanisms in apoptosis and autophagy.
Collapse
Affiliation(s)
- Rong Yu
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
| | - Urban Lendahl
- Department of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Monica Nistér
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
- *Correspondence: Monica Nistér
| | - Jian Zhao
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital Solna, Stockholm, Sweden
- Jian Zhao
| |
Collapse
|
24
|
Montava-Garriga L, Ganley IG. Outstanding Questions in Mitophagy: What We Do and Do Not Know. J Mol Biol 2020; 432:206-230. [DOI: 10.1016/j.jmb.2019.06.032] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/28/2019] [Accepted: 06/30/2019] [Indexed: 12/18/2022]
|
25
|
Fung TS, Ji WK, Higgs HN, Chakrabarti R. Two distinct actin filament populations have effects on mitochondria, with differences in stimuli and assembly factors. J Cell Sci 2019; 132:jcs234435. [PMID: 31413070 PMCID: PMC6765187 DOI: 10.1242/jcs.234435] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 08/07/2019] [Indexed: 12/12/2022] Open
Abstract
Recent studies show that mitochondria and actin filaments work together in two contexts: (1) increased cytoplasmic calcium induces cytoplasmic actin polymerization that stimulates mitochondrial fission and (2) mitochondrial depolarization causes actin assembly around mitochondria, with roles in mitophagy. It is unclear whether these two processes utilize similar actin assembly mechanisms. Here, we show that these are distinct actin assembly mechanisms in the acute phase after treatment (<10 min). Calcium-induced actin assembly is INF2 dependent and Arp2/3 complex independent, whereas depolarization-induced actin assembly is Arp2/3 complex dependent and INF2 independent. The two types of actin polymerization are morphologically distinct, with calcium-induced filaments throughout the cytosol and depolarization-induced filaments as 'clouds' around depolarized mitochondria. We have previously shown that calcium-induced actin stimulates increases in both mitochondrial calcium and recruitment of the dynamin GTPase Drp1 (also known as DNM1L). In contrast, depolarization-induced actin is temporally associated with extensive mitochondrial dynamics that do not result in mitochondrial fission, but in circularization of the inner mitochondrial membrane (IMM). These dynamics are dependent on the protease OMA1 and independent of Drp1. Actin cloud inhibition causes increased IMM circularization, suggesting that actin clouds limit these dynamics.This article has an associated First Person interview with the first author of the paper.
Collapse
Affiliation(s)
- Tak Shun Fung
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Wei-Ke Ji
- Department of Biochemistry and Molecular Biology, School of Basic Medicine and the Collaborative Innovation Center for Brain Science, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, China
| | - Henry N Higgs
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| | - Rajarshi Chakrabarti
- Department of Biochemistry and Cell Biology, Geisel School of Medicine at Dartmouth, Hanover, NH 03755, USA
| |
Collapse
|
26
|
Mitochondrial Uncoupling: A Key Controller of Biological Processes in Physiology and Diseases. Cells 2019; 8:cells8080795. [PMID: 31366145 PMCID: PMC6721602 DOI: 10.3390/cells8080795] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022] Open
Abstract
Mitochondrial uncoupling can be defined as a dissociation between mitochondrial membrane potential generation and its use for mitochondria-dependent ATP synthesis. Although this process was originally considered a mitochondrial dysfunction, the identification of UCP-1 as an endogenous physiological uncoupling protein suggests that the process could be involved in many other biological processes. In this review, we first compare the mitochondrial uncoupling agents available in term of mechanistic and non-specific effects. Proteins regulating mitochondrial uncoupling, as well as chemical compounds with uncoupling properties are discussed. Second, we summarize the most recent findings linking mitochondrial uncoupling and other cellular or biological processes, such as bulk and specific autophagy, reactive oxygen species production, protein secretion, cell death, physical exercise, metabolic adaptations in adipose tissue, and cell signaling. Finally, we show how mitochondrial uncoupling could be used to treat several human diseases, such as obesity, cardiovascular diseases, or neurological disorders.
Collapse
|
27
|
Bogetofte H, Jensen P, Ryding M, Schmidt SI, Okarmus J, Ritter L, Worm CS, Hohnholt MC, Azevedo C, Roybon L, Bak LK, Waagepetersen H, Ryan BJ, Wade-Martins R, Larsen MR, Meyer M. PARK2 Mutation Causes Metabolic Disturbances and Impaired Survival of Human iPSC-Derived Neurons. Front Cell Neurosci 2019; 13:297. [PMID: 31333417 PMCID: PMC6624735 DOI: 10.3389/fncel.2019.00297] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 06/18/2019] [Indexed: 12/22/2022] Open
Abstract
The protein parkin, encoded by the PARK2 gene, is vital for mitochondrial homeostasis, and although it has been implicated in Parkinson’s disease (PD), the disease mechanisms remain unclear. We have applied mass spectrometry-based proteomics to investigate the effects of parkin dysfunction on the mitochondrial proteome in human isogenic induced pluripotent stem cell-derived neurons with and without PARK2 knockout (KO). The proteomic analysis quantified nearly 60% of all mitochondrial proteins, 119 of which were dysregulated in neurons with PARK2 KO. The protein changes indicated disturbances in oxidative stress defense, mitochondrial respiration and morphology, cell cycle control, and cell viability. Structural and functional analyses revealed an increase in mitochondrial area and the presence of elongated mitochondria as well as impaired glycolysis and lactate-supported respiration, leading to an impaired cell survival in PARK2 KO neurons. This adds valuable insight into the effect of parkin dysfunction in human neurons and provides knowledge of disease-related pathways that can potentially be targeted for therapeutic intervention.
Collapse
Affiliation(s)
- Helle Bogetofte
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Oxford Parkinson's Disease Centre, Medical Sciences Division, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Pia Jensen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Matias Ryding
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Sissel I Schmidt
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Justyna Okarmus
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Louise Ritter
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Christina S Worm
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | - Michaela C Hohnholt
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Carla Azevedo
- Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Laurent Roybon
- Department of Experimental Medical Science, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Lasse K Bak
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Helle Waagepetersen
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Brent J Ryan
- Oxford Parkinson's Disease Centre, Medical Sciences Division, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Richard Wade-Martins
- Oxford Parkinson's Disease Centre, Medical Sciences Division, Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| | - Martin R Larsen
- Department of Biochemistry and Molecular Biology, University of Southern Denmark, Odense, Denmark
| | - Morten Meyer
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark.,Brain Research - Inter-Disciplinary Guided Excellence, University of Southern Denmark, Odense, Denmark
| |
Collapse
|
28
|
Dietz JV, Bohovych I, Viana MP, Khalimonchuk O. Proteolytic regulation of mitochondrial dynamics. Mitochondrion 2019; 49:289-304. [PMID: 31029640 DOI: 10.1016/j.mito.2019.04.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 04/19/2019] [Indexed: 12/23/2022]
Abstract
Spatiotemporal changes in the abundance, shape, and cellular localization of the mitochondrial network, also known as mitochondrial dynamics, are now widely recognized to play a key role in mitochondrial and cellular physiology as well as disease states. This process involves coordinated remodeling of the outer and inner mitochondrial membranes by conserved dynamin-like guanosine triphosphatases and their partner molecules in response to various physiological and stress stimuli. Although the core machineries that mediate fusion and partitioning of the mitochondrial network have been extensively characterized, many aspects of their function and regulation are incompletely understood and only beginning to emerge. In the present review we briefly summarize current knowledge about how the key mitochondrial dynamics-mediating factors are regulated via selective proteolysis by mitochondrial and cellular proteolytic machineries.
Collapse
Affiliation(s)
- Jonathan V Dietz
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, United States of America
| | - Iryna Bohovych
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, United States of America
| | - Martonio Ponte Viana
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, United States of America
| | - Oleh Khalimonchuk
- Department of Biochemistry, University of Nebraska, Lincoln, NE 68588, United States of America; Nebraska Redox Biology Center, University of Nebraska, Lincoln, NE 68588, United States of America; Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, United States of America.
| |
Collapse
|
29
|
Balsa E, Soustek MS, Thomas A, Cogliati S, García-Poyatos C, Martín-García E, Jedrychowski M, Gygi SP, Enriquez JA, Puigserver P. ER and Nutrient Stress Promote Assembly of Respiratory Chain Supercomplexes through the PERK-eIF2α Axis. Mol Cell 2019; 74:877-890.e6. [PMID: 31023583 DOI: 10.1016/j.molcel.2019.03.031] [Citation(s) in RCA: 221] [Impact Index Per Article: 44.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Revised: 02/20/2019] [Accepted: 03/25/2019] [Indexed: 12/27/2022]
Abstract
Endoplasmic reticulum (ER) stress and unfolded protein response are energetically challenging under nutrient stress conditions. However, the regulatory mechanisms that control the energetic demand under nutrient and ER stress are largely unknown. Here we show that ER stress and glucose deprivation stimulate mitochondrial bioenergetics and formation of respiratory supercomplexes (SCs) through protein kinase R-like ER kinase (PERK). Genetic ablation or pharmacological inhibition of PERK suppresses nutrient and ER stress-mediated increases in SC levels and reduces oxidative phosphorylation-dependent ATP production. Conversely, PERK activation augments respiratory SCs. The PERK-eIF2α-ATF4 axis increases supercomplex assembly factor 1 (SCAF1 or COX7A2L), promoting SCs and enhanced mitochondrial respiration. PERK activation is sufficient to rescue bioenergetic defects caused by complex I missense mutations derived from mitochondrial disease patients. These studies have identified an energetic communication between ER and mitochondria, with implications in cell survival and diseases associated with mitochondrial failures.
Collapse
Affiliation(s)
- Eduardo Balsa
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Meghan S Soustek
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Ajith Thomas
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Sara Cogliati
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | | | - Elena Martín-García
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain
| | - Mark Jedrychowski
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steve P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - José Antonio Enriquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; CIBERFES, Institute of Health Carlos III, Madrid 28029, Spain
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
30
|
Abstract
Mitophagy is a vital form of autophagy for selective removal of dysfunctional or redundant mitochondria. Accumulating evidence implicates elimination of dysfunctional mitochondria as a powerful means employed by autophagy to keep the immune system in check. The process of mitophagy may restrict inflammatory cytokine secretion and directly regulate mitochondrial antigen presentation and immune cell homeostasis. In this review, we describe distinctive pathways of mammalian mitophagy and highlight recent advances relevant to its function in immunity. In addition, we further discuss the direct and indirect evidence linking mitophagy to inflammation and autoimmunity underlying the pathogenesis of autoimmune diseases including inflammatory bowel diseases (IBD), systemic lupus erythematosus (SLE) and primary biliary cirrhosis (PBC).Abbreviations: AICD: activation induced cell death; AIM2: absent in melanoma 2; ALPL/HOPS: alkaline phosphatase, biomineralization associated; AMA: anti-mitochondrial antibodies; AMFR: autocrine motility factor receptor; ATG: autophagy-related; BCL2L13: BCL2 like 13; BNIP3: BCL2 interacting protein 3; BNIP3L/NIX: BCL2 interacting protein 3 like; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; CARD: caspase recruitment domain containing; CASP1: caspase 1; CD: Crohn disease; CGAS: cyclic GMP-AMP synthase; CXCL1: C-X-C motif chemokine ligand 1; DEN: diethylnitrosamine; DLAT/PDC-E2: dihydrolipoamide S-acetyltransferase; DNM1L/Drp1: dynamin 1 like; ESCRT: endosomal sorting complexes required for transport; FKBP8: FKBP prolyl isomerase 8; FUNDC1: Fun14 domain containing 1; GABARAP: GABA type A receptor-associated protein; HMGB1: high mobility group box 1; HPIV3: human parainfluenza virus type 3; IBD: inflammatory bowel diseases; IEC: intestinal epithelial cell; IFN: interferon; IL1B/IL-1β: interleukin 1 beta; iNK: invariant natural killer; IRGM: immunity related GTPase M; LIR: LC3-interacting region; LPS: lipopolysaccharide; LRRK2: leucine rich repeat kinase 2; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MARCH5: membrane associated ring-CH-type finger 5; MAVS: mitochondrial antiviral signaling protein; MDV: mitochondria-derived vesicle; MFN1: mitofusin 1; MHC: major histocompatibility complex; MIF: macrophage migration inhibitory factor; mtAP: mitochondrial antigen presentation; mtDNA: mitochondrial DNA; MTOR: mechanistic target of rapamycin kinase; mtROS: mitochondrial ROS; MUL1: mitochondrial E3 ubiquitin protein ligase 1; NBR1: NBR1 autophagy cargo receptor; NFKB/NF-ĸB: nuclear factor kappa B subunit; NK: natural killer; NLR: NOD-like receptor; NLRC4: NLR family CARD domain containing 4; NLRP3: NLR family pyrin domain containing 3; OGDH: oxoglutarate dehydrogenase; OMM: outer mitochondrial membrane; OPTN: optineurin; ox: oxidized; PARK7: Parkinsonism associated deglycase; PBC: primary biliary cirrhosis; PEX13: peroxisomal biogenesis factor 13; PHB/PHB1: prohibitin; PHB2: prohibitin 2; PIK3C3/VPS34: phosphatidylinositol 3-kinase catalytic subunit type 3; PINK1: PTEN induced kinase 1; PLEKHM1: pleckstrin homology and RUN domain containing M1; PRKN/PARK2: parkin RBR E3 ubiquitin protein ligase; RAB: member RAS oncogene family; RHEB: Ras homolog: mTORC1 binding; RIPK2: receptor interacting serine/threonine kinase 2; RLR: DDX58/RIG-I like receptor; ROS: reactive oxygen species; SBD: small bile ducts; SLC2A1/GLUT1: solute carrier family 2 member 1; SLE: systemic lupus erythematosus; SMURF1: SMAD specific E3 ubiquitin protein ligase 1; SQSTM1/p62: sequestosome 1; TAX1BP1: Tax1 binding protein 1; TCR: T cell receptor; TFAM: transcription factor A: mitochondrial; Th17: T helper 17; TLR9: toll like receptor 9; TMEM173/STING: transmembrane protein 173; TNF/TNF-α: tumor necrosis factor; Ub: ubiquitin; UC: ulcerative colitis; ULK1: unc-51 like autophagy activating kinase 1; WIPI: WD repeat domain: phosphoinositide interacting; ZFYVE1/DFCP1: zinc finger FYVE-type containing 1.
Collapse
Affiliation(s)
- Ye Xu
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai, China.,Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Digestive Disease, Shanghai, China
| | - Jun Shen
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai, China.,Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Digestive Disease, Shanghai, China
| | - Zhihua Ran
- Division of Gastroenterology and Hepatology, Key Laboratory of Gastroenterology and Hepatology, Ministry of Health, Inflammatory Bowel Disease Research Center, Shanghai, China.,Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.,Shanghai Institute of Digestive Disease, Shanghai, China
| |
Collapse
|
31
|
Sb H, X J, Qh Y, Xr Z, Bb Z, Kh W, Xy S, Yt C, Xr R, Jf M, G W, Yh P. The vicious circle between mitochondrial oxidative stress and dynamic abnormality mediates triethylene glycol dimethacrylate-induced preodontoblast apoptosis. Free Radic Biol Med 2019; 134:644-656. [PMID: 30776408 DOI: 10.1016/j.freeradbiomed.2019.02.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/13/2019] [Accepted: 02/13/2019] [Indexed: 12/13/2022]
Abstract
Oxidative stress (OS) plays crucial roles in triethylene glycol dimethacrylate (TEGDMA, a major component in dental resin)-induced apoptosis of dental pulp cells. Mitochondria are important target organelles for regulating the balance of OS, meanwhile, imbalance of the mitochondrial dynamic associated with mitochondrial dysfunction is one major molecular mechanism for oxidative damages. However, whether these mitochondrial dependent pathways were involved in the apoptosis of dental pulp cells induced by TDGDMA remains unclarified. We demonstrated that TEGDMA decreased viability and induced apoptosis of mouse preodontoblasts (mDPC6T cell line) in a time- and dose-dependent manner. Furthermore, TEGDMA elevated the mitochondrial OS status and induced mitochondrial dysfunction, as reflected by the significant decrease of mitochondrial membrane potential, ATP production, the activity of Complex III and citrate synthase. In this process, we detected a dramatically impaired mitochondrial dynamic that was reflected by significantly enhanced mitochondrial fragmentation. Consistently, we also found a significant enhancement of the key upstream regulators for mitochondrial fission, such as short form of optic atrophy 1, dynamic related protein 1 oligomer and Fission 1. The respective inhibition of mitochondrial OS or mitochondrial fission could mutually attenuate each other, thereby significantly preventing both mitochondrial dysfunction and cell apoptosis. In conclusion, TEGDMA-induced preodontoblasts apoptosis was mediated by the vicious circle between mitochondrial OS and dynamic abnormality, which represented a new target to prevent TEGDMA-induced dental pulp cells apoptosis.
Collapse
Affiliation(s)
- Huang Sb
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China; Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China; Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, 1081 LA, the Netherlands.
| | - Jin X
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China
| | - Yu Qh
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China
| | - Zhang Xr
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China
| | - Zheng Bb
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China
| | - Wang Kh
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China
| | - Sun Xy
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China
| | - Chen Yt
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China
| | - Ren Xr
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China
| | - Ma Jf
- Department of Prosthodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China; Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China
| | - Wu G
- Department of Oral Implantology and Prosthetic Dentistry, Academic Centre for Dentistry Amsterdam (ACTA), University of Amsterdam and Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, 1081 LA, the Netherlands.
| | - Pan Yh
- Institute of Stomatology, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China; Department of Endodontics, School and Hospital of Stomatology, Wenzhou Medical University, Wenzhou, PR China.
| |
Collapse
|
32
|
Alavi MV. Targeted OMA1 therapies for cancer. Int J Cancer 2019; 145:2330-2341. [PMID: 30714136 DOI: 10.1002/ijc.32177] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 01/20/2019] [Accepted: 01/23/2019] [Indexed: 12/12/2022]
Abstract
The mitochondrial inner membrane proteins OMA1 and OPA1 belong to the BAX/BAK1-dependent apoptotic signaling pathway, which can be regulated by tumor protein p53 and the prohibitins PHB and PHB2 in the context of neoplastic disease. For the most part these proteins have been studied separate from each other. Here, I argue that the OMA1 mechanism of action represents the missing link between p53 and cytochrome c release. The mitochondrial fusion protein OPA1 is cleaved by OMA1 in a stress-dependent manner generating S-OPA1. Excessive S-OPA1 can facilitate outer membrane permeabilization upon BAX/BAK1 activation through its membrane shaping properties. p53 helps outer membrane permeabilization in a 2-step process. First, cytosolic p53 activates BAX/BAK1 at the mitochondrial surface. Then, in a second step, p53 binds to prohibitin thereby releasing the restraint on OMA1. This activates OMA1, which cleaves OPA1 and promotes cytochrome c release. Clearly, OMA1 and OPA1 are not root causes for cancer. Yet many cancer cells rely on this pathway for survival, which can explain why loss of p53 function promotes tumor growth and confers resistance to chemotherapies.
Collapse
|
33
|
Lee H, Yoon Y. Mitochondrial Membrane Dynamics-Functional Positioning of OPA1. Antioxidants (Basel) 2018; 7:antiox7120186. [PMID: 30544804 PMCID: PMC6316456 DOI: 10.3390/antiox7120186] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 12/04/2018] [Accepted: 12/06/2018] [Indexed: 12/21/2022] Open
Abstract
The maintenance of mitochondrial energetics requires the proper regulation of mitochondrial morphology, and vice versa. Mitochondrial dynamins control mitochondrial morphology by mediating fission and fusion. One of them, optic atrophy 1 (OPA1), is the mitochondrial inner membrane remodeling protein. OPA1 has a dual role in maintaining mitochondrial morphology and energetics through mediating inner membrane fusion and maintaining the cristae structure. OPA1 is expressed in multiple variant forms through alternative splicing and post-translational proteolytic cleavage, but the functional differences between these variants have not been completely understood. Recent studies generated new information regarding the role of OPA1 cleavage. In this review, we will first provide a brief overview of mitochondrial membrane dynamics by describing fission and fusion that are mediated by mitochondrial dynamins. The second part describes OPA1-mediated fusion and energetic maintenance, the role of OPA1 cleavage, and a new development in OPA1 function, in which we will provide new insight for what OPA1 does and what proteolytic cleavage of OPA1 is for.
Collapse
Affiliation(s)
- Hakjoo Lee
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA.
| |
Collapse
|
34
|
Cadete VJJ, Vasam G, Menzies KJ, Burelle Y. Mitochondrial quality control in the cardiac system: An integrative view. Biochim Biophys Acta Mol Basis Dis 2018; 1865:782-796. [PMID: 30472159 DOI: 10.1016/j.bbadis.2018.11.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/29/2018] [Accepted: 11/15/2018] [Indexed: 01/26/2023]
Abstract
Recent studies have led to the discovery of multiple mitochondrial quality control (mQC) processes that operate at various scales, ranging from the degradation of proteins by mitochondrial proteases to the degradation of selected cargos or entire organelles in lysosomes. While the mechanisms governing these mQC processes are progressively being delineated, their role and importance remain unclear. Converging evidence however point to a complex system whereby multiple and partly overlapping processes are recruited to orchestrate a cell type specific mQC response that is adapted to the physiological state and level of stress encountered. Knowledge gained from basic model systems of mQC therefore need to be integrated within organ-specific (patho)physiological frameworks. Building on this notion, this article focuses on mQC in the heart, where developmental metabolic reprogramming, sustained contraction, and multiple pathophysiological conditions pose broadly different constraints. We provide an overview of current knowledge of mQC processes, and discuss their implication in cardiac mQC under normal and diseased conditions.
Collapse
Affiliation(s)
- Virgilio J J Cadete
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Goutham Vasam
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Keir J Menzies
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa Brain and Mind Research Institute and Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON, Canada
| | - Yan Burelle
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
35
|
Abstract
Mitochondria undergo continuous challenges in the course of their life, from their generation to their degradation. These challenges include the management of reactive oxygen species, the proper assembly of mitochondrial respiratory complexes and the need to balance potential mutations in the mitochondrial DNA. The detection of damage and the ability to keep it under control is critical to fine-tune mitochondrial function to the organismal energy needs. In this review, we will analyze the multiple mechanisms that safeguard mitochondrial function in light of in crescendo damage. This sequence of events will include initial defense against excessive reactive oxygen species production, compensation mechanisms by the unfolded protein response (UPRmt), mitochondrial dynamics and elimination by mitophagy.
Collapse
Affiliation(s)
- Miriam Valera-Alberni
- Nestlé Institute of Health Sciences (NIHS), EPFL Innovation Park, 1015 Lausanne.,School of Life Sciences, EPFL, 1015 Lausanne
| | - Carles Canto
- Nestlé Institute of Health Sciences (NIHS), EPFL Innovation Park, 1015 Lausanne.,School of Life Sciences, EPFL, 1015 Lausanne
| |
Collapse
|
36
|
Vandekeere S, Dubois C, Kalucka J, Sullivan MR, García-Caballero M, Goveia J, Chen R, Diehl FF, Bar-Lev L, Souffreau J, Pircher A, Kumar S, Vinckier S, Hirabayashi Y, Furuya S, Schoonjans L, Eelen G, Ghesquière B, Keshet E, Li X, Vander Heiden MG, Dewerchin M, Carmeliet P. Serine Synthesis via PHGDH Is Essential for Heme Production in Endothelial Cells. Cell Metab 2018; 28:573-587.e13. [PMID: 30017355 DOI: 10.1016/j.cmet.2018.06.009] [Citation(s) in RCA: 126] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 04/04/2018] [Accepted: 06/14/2018] [Indexed: 01/09/2023]
Abstract
The role of phosphoglycerate dehydrogenase (PHGDH), a key enzyme of the serine synthesis pathway (SSP), in endothelial cells (ECs) remains poorly characterized. We report that mouse neonates with EC-specific PHGDH deficiency suffer lethal vascular defects within days of gene inactivation, due to reduced EC proliferation and survival. In addition to nucleotide synthesis impairment, PHGDH knockdown (PHGDHKD) caused oxidative stress, due not only to decreased glutathione and NADPH synthesis but also to mitochondrial dysfunction. Electron transport chain (ETC) enzyme activities were compromised upon PHGDHKD because of insufficient heme production due to cellular serine depletion, not observed in other cell types. As a result of heme depletion, elevated reactive oxygen species levels caused EC demise. Supplementation of hemin in PHGDHKD ECs restored ETC function and rescued the apoptosis and angiogenesis defects. These data argue that ECs die upon PHGDH inhibition, even without external serine deprivation, illustrating an unusual importance of serine synthesis for ECs.
Collapse
Affiliation(s)
- Saar Vandekeere
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Charlotte Dubois
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Joanna Kalucka
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Mark R Sullivan
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Melissa García-Caballero
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Jermaine Goveia
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Rongyuan Chen
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China
| | - Frances F Diehl
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Libat Bar-Lev
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem 91120, Israel
| | - Joris Souffreau
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Andreas Pircher
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Saran Kumar
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem 91120, Israel
| | - Stefan Vinckier
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Yoshio Hirabayashi
- Laboratory for Molecular Membrane Neuroscience, RIKEN Brain Science Institute, Wako City, Saimata 351-0198, Japan
| | - Shigeki Furuya
- Department of Bioscience and Biotechnology, Kyushu University, Fukuoka 812-8581, Japan
| | - Luc Schoonjans
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Guy Eelen
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Bart Ghesquière
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium
| | - Eli Keshet
- Department of Developmental Biology and Cancer Research, The Hebrew University, Jerusalem 91120, Israel
| | - Xuri Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China.
| | - Matthew G Vander Heiden
- The Koch Institute for Integrative Cancer Research and Department of Biology, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Dana-Farber Cancer Institute, Boston, MA 02115, USA
| | - Mieke Dewerchin
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium.
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, VIB Center for Cancer Biology, VIB, Leuven 3000, Belgium; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-Sen University, Guangzhou 510060, Guangdong, P.R. China; Laboratory of Angiogenesis and Vascular Metabolism, VIB-KU Leuven Center for Cancer Biology, Department of Oncology, KU Leuven, Leuven 3000, Belgium.
| |
Collapse
|
37
|
Zhang T, Wu P, Zhang JH, Li Y, Xu S, Wang C, Wang L, Zhang G, Dai J, Zhu S, Liu Y, Liu B, Reis C, Shi H. Docosahexaenoic Acid Alleviates Oxidative Stress-Based Apoptosis Via Improving Mitochondrial Dynamics in Early Brain Injury After Subarachnoid Hemorrhage. Cell Mol Neurobiol 2018; 38:1413-1423. [PMID: 30084007 DOI: 10.1007/s10571-018-0608-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 08/01/2018] [Indexed: 01/04/2023]
Abstract
Mitochondrial dysfunction is considered a crucial therapeutic target for early brain injury following subarachnoid hemorrhage (SAH). Emerging evidence indicates that docosahexaenoic acid (DHA), an essential omega-3 fatty acid, protects mitochondria in various chronic diseases. This study aimed to investigate the neuroprotective effects of DHA on mitochondrial dynamic dysfunction after EBI using in vivo and in vitro approaches. For in vivo experiments, the rat endovascular perforation SAH model was performed, whereby DHA was administered intravenously 1 h after induction of SAH. Primary cultured neurons treated with oxyhemoglobin (OxyHb) for 24 h were used to mimic SAH in vitro. Our results demonstrated that DHA improved neurological deficits and reduced brain edema in rats with SAH, and attenuated OxyHb-induced neuronal death in primary cultured cells. DHA reduced the amount of reactive oxygen species-positive cells and improved cell viability when compared to the SAH + vehicle group in vitro. DHA attenuated malondialdehyde levels and superoxide dismutase stress, increased Bcl2 and Bcl-xl, and decreased Bax and cleaved caspase-3 in vivo. Additionally, DHA ameliorated mitochondrial dysfunction, upregulated the mitochondrial fusion-related protein Optic Atrophy 1, and downregulated the mitochondrial fission-related protein Dynamin-Related-Protein 1 (Drp1) and Serine 616 phosphorylated Drp1 after SAH both in vitro and in vivo. Taken together, our current study demonstrates that DHA might prevent oxidative stress-based apoptosis after SAH. The characterization of the underlying molecular mechanisms may further improve mitochondrial dynamics-related signaling pathways.
Collapse
Affiliation(s)
- Tongyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Pei Wu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - John H Zhang
- Departments of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Yuchen Li
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shancai Xu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Chunlei Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Ligang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Guang Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jiaxing Dai
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shiyi Zhu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yao Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Binbing Liu
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Cesar Reis
- Departments of Physiology and Pharmacology, Loma Linda University, Loma Linda, CA, USA
| | - Huaizhang Shi
- Department of Neurosurgery, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China.
| |
Collapse
|
38
|
A Disturbance in the Force: Cellular Stress Sensing by the Mitochondrial Network. Antioxidants (Basel) 2018; 7:antiox7100126. [PMID: 30249006 PMCID: PMC6211095 DOI: 10.3390/antiox7100126] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Revised: 09/13/2018] [Accepted: 09/17/2018] [Indexed: 12/15/2022] Open
Abstract
As a highly dynamic organellar network, mitochondria are maintained as an organellar network by delicately balancing fission and fusion pathways. This homeostatic balance of organellar dynamics is increasingly revealed to play an integral role in sensing cellular stress stimuli. Mitochondrial fission/fusion balance is highly sensitive to perturbations such as loss of bioenergetic function, oxidative stress, and other stimuli, with mechanistic contribution to subsequent cell-wide cascades including inflammation, autophagy, and apoptosis. The overlapping activity with m-AAA protease 1 (OMA1) metallopeptidase, a stress-sensitive modulator of mitochondrial fusion, and dynamin-related protein 1 (DRP1), a regulator of mitochondrial fission, are key factors that shape mitochondrial dynamics in response to various stimuli. As such, OMA1 and DRP1 are critical factors that mediate mitochondrial roles in cellular stress-response signaling. Here, we explore the current understanding and emerging questions in the role of mitochondrial dynamics in sensing cellular stress as a dynamic, responsive organellar network.
Collapse
|
39
|
Lang A, Anand R, Altinoluk-Hambüchen S, Ezzahoini H, Stefanski A, Iram A, Bergmann L, Urbach J, Böhler P, Hänsel J, Franke M, Stühler K, Krutmann J, Scheller J, Stork B, Reichert AS, Piekorz RP. SIRT4 interacts with OPA1 and regulates mitochondrial quality control and mitophagy. Aging (Albany NY) 2018; 9:2163-2189. [PMID: 29081403 PMCID: PMC5680561 DOI: 10.18632/aging.101307] [Citation(s) in RCA: 98] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Accepted: 10/15/2017] [Indexed: 12/13/2022]
Abstract
The stress-responsive mitochondrial sirtuin SIRT4 controls cellular energy metabolism in a NAD+-dependent manner and is implicated in cellular senescence and aging. Here we reveal a novel function of SIRT4 in mitochondrial morphology/quality control and regulation of mitophagy. We report that moderate overexpression of SIRT4, but not its enzymatically inactive mutant H161Y, sensitized cells to mitochondrial stress. CCCP-triggered dissipation of the mitochondrial membrane potential resulted in increased mitochondrial ROS levels and autophagic flux, but surprisingly led to increased mitochondrial mass and decreased Parkin-regulated mitophagy. The anti-respiratory effect of elevated SIRT4 was accompanied by increased levels of the inner-membrane bound long form of the GTPase OPA1 (L-OPA1) that promotes mitochondrial fusion and thereby counteracts fission and mitophagy. Consistent with this, upregulation of endogenous SIRT4 expression in fibroblast models of senescence either by transfection with miR-15b inhibitors or by ionizing radiation increased L-OPA1 levels and mitochondrial fusion in a SIRT4-dependent manner. We further demonstrate that SIRT4 interacts physically with OPA1 in co-immunoprecipitation experiments. Overall, we propose that the SIRT4-OPA1 axis is causally linked to mitochondrial dysfunction and altered mitochondrial dynamics that translates into aging-associated decreased mitophagy based on an unbalanced mitochondrial fusion/fission cycle.
Collapse
Affiliation(s)
- Alexander Lang
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Ruchika Anand
- Institut für Biochemie und Molekularbiologie I, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Simone Altinoluk-Hambüchen
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Hakima Ezzahoini
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Anja Stefanski
- Molecular Proteomics Laboratory (BMFZ), Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Afshin Iram
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Laura Bergmann
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jennifer Urbach
- Institut für Biochemie und Molekularbiologie I, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Philip Böhler
- Institut für Molekulare Medizin I, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jan Hänsel
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Manuel Franke
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Kai Stühler
- Molecular Proteomics Laboratory (BMFZ), Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jean Krutmann
- IUF - Leibniz Institut für Umweltmedizinische Forschung, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Jürgen Scheller
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Björn Stork
- Institut für Molekulare Medizin I, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Andreas S Reichert
- Institut für Biochemie und Molekularbiologie I, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Roland P Piekorz
- Institut für Biochemie und Molekularbiologie II, Medizinische Fakultät der Heinrich-Heine-Universität, Düsseldorf, Germany
| |
Collapse
|
40
|
Marcassa E, Kallinos A, Jardine J, Rusilowicz-Jones EV, Martinez A, Kuehl S, Islinger M, Clague MJ, Urbé S. Dual role of USP30 in controlling basal pexophagy and mitophagy. EMBO Rep 2018; 19:embr.201745595. [PMID: 29895712 PMCID: PMC6030704 DOI: 10.15252/embr.201745595] [Citation(s) in RCA: 109] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 05/15/2018] [Accepted: 05/18/2018] [Indexed: 12/20/2022] Open
Abstract
USP30 is an integral protein of the outer mitochondrial membrane that counteracts PINK1 and Parkin‐dependent mitophagy following acute mitochondrial depolarisation. Here, we use two distinct mitophagy reporter systems to reveal tonic suppression by USP30, of a PINK1‐dependent component of basal mitophagy in cells lacking detectable Parkin. We propose that USP30 acts upstream of PINK1 through modulation of PINK1‐substrate availability and thereby determines the potential for mitophagy initiation. We further show that a fraction of endogenous USP30 is independently targeted to peroxisomes where it regulates basal pexophagy in a PINK1‐ and Parkin‐independent manner. Thus, we reveal a critical role of USP30 in the clearance of the two major sources of ROS in mammalian cells and in the regulation of both a PINK1‐dependent and a PINK1‐independent selective autophagy pathway.
Collapse
Affiliation(s)
- Elena Marcassa
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Andreas Kallinos
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Jane Jardine
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Emma V Rusilowicz-Jones
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Aitor Martinez
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Sandra Kuehl
- Institute of Neuroanatomy, Centre for Biomedicine and Medical Technology Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Markus Islinger
- Institute of Neuroanatomy, Centre for Biomedicine and Medical Technology Mannheim, Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany
| | - Michael J Clague
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Sylvie Urbé
- Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| |
Collapse
|
41
|
Rodger CE, McWilliams TG, Ganley IG. Mammalian mitophagy - from in vitro molecules to in vivo models. FEBS J 2018; 285:1185-1202. [PMID: 29151277 PMCID: PMC5947125 DOI: 10.1111/febs.14336] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 10/09/2017] [Accepted: 11/14/2017] [Indexed: 12/25/2022]
Abstract
The autophagic turnover of mitochondria, termed mitophagy, is thought to play an essential role in not only maintaining the health of the mitochondrial network but also that of the cell and organism as a whole. We have come a long way in identifying the molecular components required for mitophagy through extensive in vitro work and cell line characterisation, yet the physiological significance and context of these pathways remain largely unexplored. This is highlighted by the recent development of new mouse models that have revealed a striking level of variation in mitophagy, even under normal conditions. Here, we focus on programmed mitophagy and summarise our current understanding of why, how and where this takes place in mammals.
Collapse
Affiliation(s)
- Catherine E Rodger
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, UK
| | - Thomas G McWilliams
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, UK
| | - Ian G Ganley
- MRC Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, UK
| |
Collapse
|
42
|
Acin-Perez R, Lechuga-Vieco AV, del Mar Muñoz M, Nieto-Arellano R, Torroja C, Sánchez-Cabo F, Jiménez C, González-Guerra A, Carrascoso I, Benincá C, Quiros PM, López-Otín C, Castellano JM, Ruíz-Cabello J, Jiménez-Borreguero LJ, Enríquez JA. Ablation of the stress protease OMA1 protects against heart failure in mice. Sci Transl Med 2018; 10:10/434/eaan4935. [DOI: 10.1126/scitranslmed.aan4935] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 09/14/2017] [Accepted: 02/13/2018] [Indexed: 12/14/2022]
|
43
|
Schlattner U, Tokarska-Schlattner M, Epand RM, Boissan M, Lacombe ML, Kagan VE. NME4/nucleoside diphosphate kinase D in cardiolipin signaling and mitophagy. J Transl Med 2018. [PMID: 29035377 DOI: 10.38/labinvest.2017.113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023] Open
Abstract
Mitophagy is an emerging paradigm for mitochondrial quality control and cell homeostasis. Dysregulation of mitophagy can lead to human pathologies such as neurodegenerative disorders and contributes to the aging process. Complex protein signaling cascades have been described that regulate mitophagy. We have identified a novel lipid signaling pathway that involves the phospholipid cardiolipin (CL). CL is synthesized and normally confined at the inner mitochondrial membrane. However, upon a mitophagic trigger, ie, collapse of the inner membrane potential, CL is rapidly externalized to the mitochondrial surface with the assistance of the hexameric nucleoside diphosphate kinase D (NME4, NDPK-D, or NM23-H4). In addition to its NDP kinase activity, NME4/NDPK-D shows intermembrane phospholipid transfer activity in vitro and in cellular systems, which relies on NME4/NDPK-D interaction with CL, CL-dependent crosslinking of inner and outer mitochondrial membranes by symmetrical, hexameric NME4/NDPK-D, and a putative NME4/NDPK-D-based CL-transfer pathway. CL exposed at the mitochondrial surface then serves as an 'eat me' signal for the mitophagic machinery; it is recognized by the LC3 receptor of autophagosomes, targeting the dysfunctional mitochondrion to lysosomal degradation. Similar NME4-supported CL externalization is likely also involved in apoptosis and inflammatory reactions.
Collapse
Affiliation(s)
- Uwe Schlattner
- University Grenoble Alpes, Inserm, Laboratory of Fundamental and Applied Bioenergetics (LBFA) and SFR Environmental and Systems Biology (BEeSy), Grenoble, France
| | - Malgorzata Tokarska-Schlattner
- University Grenoble Alpes, Inserm, Laboratory of Fundamental and Applied Bioenergetics (LBFA) and SFR Environmental and Systems Biology (BEeSy), Grenoble, France
| | - Richard M Epand
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON, Canada
| | - Mathieu Boissan
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, UMRS938, Saint-Antoine Research Center, Paris, France
- Hôpitaux Universitaires Est Parisien-Tenon, Assistance Publique Hôpitaux de Paris, Laboratoire de Biochimie et Hormonologie, Paris, France
| | - Marie-Lise Lacombe
- Sorbonne Universités, UPMC Univ Paris 06, Inserm, UMRS938, Saint-Antoine Research Center, Paris, France
| | - Valerian E Kagan
- Center for Free Radical and Antioxidant Health, Department of Environmental Health, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
44
|
NME4/nucleoside diphosphate kinase D in cardiolipin signaling and mitophagy. J Transl Med 2018; 98:228-232. [PMID: 29035377 DOI: 10.1038/labinvest.2017.113] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/09/2017] [Accepted: 08/10/2017] [Indexed: 01/12/2023] Open
Abstract
Mitophagy is an emerging paradigm for mitochondrial quality control and cell homeostasis. Dysregulation of mitophagy can lead to human pathologies such as neurodegenerative disorders and contributes to the aging process. Complex protein signaling cascades have been described that regulate mitophagy. We have identified a novel lipid signaling pathway that involves the phospholipid cardiolipin (CL). CL is synthesized and normally confined at the inner mitochondrial membrane. However, upon a mitophagic trigger, ie, collapse of the inner membrane potential, CL is rapidly externalized to the mitochondrial surface with the assistance of the hexameric nucleoside diphosphate kinase D (NME4, NDPK-D, or NM23-H4). In addition to its NDP kinase activity, NME4/NDPK-D shows intermembrane phospholipid transfer activity in vitro and in cellular systems, which relies on NME4/NDPK-D interaction with CL, CL-dependent crosslinking of inner and outer mitochondrial membranes by symmetrical, hexameric NME4/NDPK-D, and a putative NME4/NDPK-D-based CL-transfer pathway. CL exposed at the mitochondrial surface then serves as an 'eat me' signal for the mitophagic machinery; it is recognized by the LC3 receptor of autophagosomes, targeting the dysfunctional mitochondrion to lysosomal degradation. Similar NME4-supported CL externalization is likely also involved in apoptosis and inflammatory reactions.
Collapse
|
45
|
Bottega R, Nicchia E, Cappelli E, Ravera S, De Rocco D, Faleschini M, Corsolini F, Pierri F, Calvillo M, Russo G, Casazza G, Ramenghi U, Farruggia P, Dufour C, Savoia A. Hypomorphic FANCA mutations correlate with mild mitochondrial and clinical phenotype in Fanconi anemia. Haematologica 2017; 103:417-426. [PMID: 29269525 PMCID: PMC5830397 DOI: 10.3324/haematol.2017.176131] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/14/2017] [Indexed: 11/16/2022] Open
Abstract
Fanconi anemia is a rare disease characterized by congenital malformations, aplastic anemia, and predisposition to cancer. Despite the consolidated role of the Fanconi anemia proteins in DNA repair, their involvement in mitochondrial function is emerging. The purpose of this work was to assess whether the mitochondrial phenotype, independent of genomic integrity, could correlate with patient phenotype. We evaluated mitochondrial and clinical features of 11 affected individuals homozygous or compound heterozygous for p.His913Pro and p.Arg951Gln/Trp, the two residues of FANCA that are more frequently affected in our cohort of patients. Although p.His913Pro and p.Arg951Gln proteins are stably expressed in cytoplasm, they are unable to migrate in the nucleus, preventing cells from repairing DNA. In these cells, the electron transfer between respiring complex I–III is reduced and the ATP/AMP ratio is impaired with defective ATP production and AMP accumulation. These activities are intermediate between those observed in wild-type and FANCA−/− cells, suggesting that the variants at residues His913 and Arg951 are hypomorphic mutations. Consistent with these findings, the clinical phenotype of most of the patients carrying these mutations is mild. These data further support the recent finding that the Fanconi anemia proteins play a role in mitochondria, and open up possibilities for genotype/phenotype studies based on novel mitochondrial criteria.
Collapse
Affiliation(s)
- Roberta Bottega
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Elena Nicchia
- Department of Medical Sciences, University of Trieste, Genoa, Italy
| | - Enrico Cappelli
- Clinical and Experimental Hematology Unit, "G. Gaslini" Children's Hospital, Genoa, Italy
| | - Silvia Ravera
- Department of Pharmacy (DIFAR), Biochemistry Lab, University of Genoa, Genoa, Italy
| | - Daniela De Rocco
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Michela Faleschini
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy
| | - Fabio Corsolini
- U.O.S.D. Centro di Diagnostica Genetica e Biochimica delle Malattie Metaboliche, "G. Gaslini" Children's Hospital, Genoa, Italy
| | - Filomena Pierri
- Clinical and Experimental Hematology Unit, "G. Gaslini" Children's Hospital, Genoa, Italy
| | - Michaela Calvillo
- Clinical and Experimental Hematology Unit, "G. Gaslini" Children's Hospital, Genoa, Italy
| | - Giovanna Russo
- Oncology Hematology Pediatric Unit, "Policlinico - Vittorio Emanuele", University of Catania, Pisa, Italy
| | - Gabriella Casazza
- Pediatric Onco-Hematology, Azienda Ospedaliera/Universitaria Pisana, Pisa, Italy
| | - Ugo Ramenghi
- Department of Pediatric and Public Health Sciences, University of Torino, Palermo, Italy
| | - Piero Farruggia
- Pediatric Onco-Hematology, ARNAS Civico Hospital, Palermo, Italy
| | - Carlo Dufour
- Clinical and Experimental Hematology Unit, "G. Gaslini" Children's Hospital, Genoa, Italy
| | - Anna Savoia
- Institute for Maternal and Child Health - IRCCS "Burlo Garofolo", Trieste, Italy .,Department of Medical Sciences, University of Trieste, Genoa, Italy
| |
Collapse
|
46
|
Cho I, Song HO, Cho JH. Mitochondrial Uncoupling Attenuates Age-Dependent Neurodegeneration in C. elegans. Mol Cells 2017; 40:864-870. [PMID: 29081084 PMCID: PMC5712516 DOI: 10.14348/molcells.2017.0172] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 09/08/2017] [Accepted: 09/21/2017] [Indexed: 12/19/2022] Open
Abstract
The uncoupling protein 4 (ucp-4) gene is involved in age-dependent neurodegeneration in C. elegans. Therefore, we aimed to investigate the mechanism underlying the association between mitochondrial uncoupling and neurodegeneration by examining the effects of uncoupling agents and ucp-4 overexpression in C. elegans. Treatment with either DNP or CCCP improved neuronal defects in wild type during aging. Uncoupling agents also restored neuronal phenotypes of ucp-4 mutants to those exhibited by wild type, while ucp-4 overexpression attenuated the severity of age-dependent neurodegeneration. Neuronal improvements were further associated with reductions in mitochondrial membrane potentials. However, these age-dependent neuroprotective effects were limited in mitophagy-deficient mutant, pink-1, background. These results suggest that membrane uncoupling can attenuate age-dependent neurodegeneration by stimulating mitophagy.
Collapse
Affiliation(s)
- Injeong Cho
- Department of Biology Education, College of Education, Chosun University, Gwangju 61452,
Korea
| | - Hyun-Ok Song
- Department of Infection Biology, Wonkwang University School of Medicine, Iksan 54538,
Korea
| | - Jeong Hoon Cho
- Department of Biology Education, College of Education, Chosun University, Gwangju 61452,
Korea
| |
Collapse
|
47
|
Le Guerroué F, Eck F, Jung J, Starzetz T, Mittelbronn M, Kaulich M, Behrends C. Autophagosomal Content Profiling Reveals an LC3C-Dependent Piecemeal Mitophagy Pathway. Mol Cell 2017; 68:786-796.e6. [DOI: 10.1016/j.molcel.2017.10.029] [Citation(s) in RCA: 65] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Revised: 08/01/2017] [Accepted: 10/20/2017] [Indexed: 10/18/2022]
|
48
|
Chang JC, Hoel F, Liu KH, Wei YH, Cheng FC, Kuo SJ, Tronstad KJ, Liu CS. Peptide-mediated delivery of donor mitochondria improves mitochondrial function and cell viability in human cybrid cells with the MELAS A3243G mutation. Sci Rep 2017; 7:10710. [PMID: 28878349 PMCID: PMC5587702 DOI: 10.1038/s41598-017-10870-5] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Accepted: 08/16/2017] [Indexed: 02/07/2023] Open
Abstract
The cell penetrating peptide, Pep-1, has been shown to facilitate cellular uptake of foreign mitochondria but further research is required to evaluate the use of Pep-1-mediated mitochondrial delivery (PMD) in treating mitochondrial defects. Presently, we sought to determine whether mitochondrial transplantation rescue mitochondrial function in a cybrid cell model of mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS) disease. Following PMD, recipient cells had internalized donor mitochondria after 1 h, and expressed higher levels of normal mitochondrial DNA, particularly at the end of the treatment and 11 days later. After 4 days, mitochondrial respiratory function had recovered and biogenesis was evident in the Pep-1 and PMD groups, compared to the untreated MELAS group. However, only PMD was able to reverse the fusion-to-fission ratio of mitochondrial morphology, and mitochondria shaping proteins resembled the normal pattern seen in the control group. Cell survival following hydrogen peroxide-induced oxidative stress was also improved in the PMD group. Finally, we observed that PMD partially normalized cytokine expression, including that of interleukin (IL)-7, granulocyte macrophage–colony-stimulating factor (GM-CSF), and vascular endothelial growth factor (VEGF), in the MELAS cells. Presently, our data further confirm the protective effects of PMD as well in MELAS disease.
Collapse
Affiliation(s)
- Jui-Chih Chang
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Fredrik Hoel
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Ko-Hung Liu
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan
| | - Yau-Huei Wei
- Department of Biochemistry and Molecular Biology, School of Life Sciences, National Yang-Ming University, Taipei, Taiwan.,Department of Medicine, Mackay Medical College, Taipei, Taiwan
| | - Fu-Chou Cheng
- Stem Cell Center, Department of Medical Research, Taichung Veterans General Hospital, Changhua, Taiwan
| | - Shou-Jen Kuo
- Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
| | | | - Chin-San Liu
- Vascular and Genomic Center, Changhua Christian Hospital, Changhua, Taiwan. .,Department of Neurology, Changhua Christian Hospital, Changhua, Taiwan.
| |
Collapse
|
49
|
Mitochondrial fission and fusion. Biochem Soc Trans 2017; 44:1725-1735. [PMID: 27913683 DOI: 10.1042/bst20160129] [Citation(s) in RCA: 119] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2016] [Revised: 09/15/2016] [Accepted: 10/05/2016] [Indexed: 01/27/2023]
Abstract
Mitochondrial fission and fusion have been recognized as critical processes in the health of mitochondria and cells. Two decades of studies have generated a great deal of information about mitochondrial fission and fusion; however, still much needs to be understood for the basic molecular mechanisms of these important cellular processes. The core protein factors for mitochondrial fission and fusion are dynamin proteins that possess membrane-remodeling properties. This short review covers a recent development and understanding of the mechanisms by which these mechanochemical enzymes mediate mitochondrial fission and fusion.
Collapse
|
50
|
Constriction of the mitochondrial inner compartment is a priming event for mitochondrial division. Nat Commun 2017; 8:15754. [PMID: 28598422 PMCID: PMC5472732 DOI: 10.1038/ncomms15754] [Citation(s) in RCA: 115] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2016] [Accepted: 04/26/2017] [Indexed: 02/02/2023] Open
Abstract
Mitochondrial division is critical for the maintenance and regulation of mitochondrial function, quality and distribution. This process is controlled by cytosolic actin-based constriction machinery and dynamin-related protein 1 (Drp1) on mitochondrial outer membrane (OMM). Although mitochondrial physiology, including oxidative phosphorylation, is also important for efficient mitochondrial division, morphological alterations of the mitochondrial inner-membrane (IMM) have not been clearly elucidated. Here we report spontaneous and repetitive constriction of mitochondrial inner compartment (CoMIC) associated with subsequent division in neurons. Although CoMIC is potentiated by inhibition of Drp1 and occurs at the potential division spots contacting the endoplasmic reticulum, it appears on IMM independently of OMM. Intra-mitochondrial influx of Ca2+ induces and potentiates CoMIC, and leads to K+-mediated mitochondrial bulging and depolarization. Synergistically, optic atrophy 1 (Opa1) also regulates CoMIC via controlling Mic60-mediated OMM-IMM tethering. Therefore, we propose that CoMIC is a priming event for efficient mitochondrial division.
Collapse
|