1
|
Feng L, Li B, Yong SS, Wen X, Tian Z. The emerging role of exercise in Alzheimer's disease: Focus on mitochondrial function. Ageing Res Rev 2024; 101:102486. [PMID: 39243893 DOI: 10.1016/j.arr.2024.102486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 08/31/2024] [Indexed: 09/09/2024]
Abstract
Alzheimer's disease (AD) is an age-related neurodegenerative disease characterized by memory impairment and cognitive dysfunction, which eventually leads to the disability and mortality of older adults. Although the precise mechanisms by which age promotes the development of AD remains poorly understood, mitochondrial dysfunction plays a central role in the development of AD. Currently, there is no effective treatment for this debilitating disease. It is well accepted that exercise exerts neuroprotective effects by ameliorating mitochondrial dysfunction in the neurons of AD, which involves multiple mechanisms, including mitochondrial dynamics, biogenesis, mitophagy, transport, and signal transduction. In addition, exercise promotes mitochondria communication with other organelles in AD neurons, which should receive more attentions in the future.
Collapse
Affiliation(s)
- Lili Feng
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China.
| | - Bowen Li
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China
| | - Su Sean Yong
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China
| | - Xu Wen
- Department of Sports Science, College of Education, Zhejiang University, Hangzhou 310030, China.
| | - Zhenjun Tian
- Institute of Sports Biology, College of Physical Education, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
2
|
Zaninello M, Baptista P, Duarte FV. Mitochondrial Dynamics and mRNA Translation: A Local Synaptic Tale. BIOLOGY 2024; 13:746. [PMID: 39336173 PMCID: PMC11428642 DOI: 10.3390/biology13090746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024]
Abstract
Mitochondria are dynamic organelles that can adjust and respond to different stimuli within a cell. This plastic ability allows them to effectively coordinate several cellular functions in cells and becomes particularly relevant in highly complex cells such as neurons. An imbalance in mitochondrial dynamics can disrupt mitochondrial function, leading to abnormal cellular function and ultimately to a range of diseases, including neurodegenerative disorders. Regulation of mRNA transport and local translation inside neurons is crucial for maintaining the proteome of distal mitochondria, which is vital for energy production and synaptic function. A significant portion of the axonal transcriptome is dedicated to mRNAs for mitochondrial proteins, emphasizing the importance of local translation in sustaining mitochondrial function in areas far from the cell body. In neurons, local translation and the regulation of mRNAs encoding mitochondrial-shaping proteins could be essential for synaptic plasticity and neuronal health. The dynamics of these mRNAs, including their transport and local translation, may influence the morphology and function of mitochondria, thereby affecting the overall energy status and responsiveness of synapses. Comprehending the mitochondria-related mRNA regulation and local translation, as well as its influence on mitochondrial morphology near the synapses will help to better understand neuronal physiology and neurological diseases where mitochondrial dysfunction and impaired synaptic plasticity play a central role.
Collapse
Affiliation(s)
- Marta Zaninello
- Institute for Genetics, University of Cologne, 50931 Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), 50931 Cologne, Germany
| | - Pedro Baptista
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Filipe V Duarte
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal
- Department of Life Sciences, University of Coimbra, 3004-504 Coimbra, Portugal
| |
Collapse
|
3
|
Lacombe A, Scorrano L. The interplay between mitochondrial dynamics and autophagy: From a key homeostatic mechanism to a driver of pathology. Semin Cell Dev Biol 2024; 161-162:1-19. [PMID: 38430721 DOI: 10.1016/j.semcdb.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 02/06/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024]
Abstract
The complex relationship between mitochondrial dynamics and autophagy illustrates how two cellular housekeeping processes are intimately linked, illuminating fundamental principles of cellular homeostasis and shedding light on disparate pathological conditions including several neurodegenerative disorders. Here we review the basic tenets of mitochondrial dynamics i.e., the concerted balance between fusion and fission of the organelle, and its interplay with macroautophagy and selective mitochondrial autophagy, also dubbed mitophagy, in the maintenance of mitochondrial quality control and ultimately in cell viability. We illustrate how conditions of altered mitochondrial dynamics reverberate on autophagy and vice versa. Finally, we illustrate how altered interplay between these two key cellular processes participates in the pathogenesis of human disorders affecting multiple organs and systems.
Collapse
Affiliation(s)
- Alice Lacombe
- Dept. of Biology, University of Padova, Padova, Italy
| | - Luca Scorrano
- Dept. of Biology, University of Padova, Padova, Italy; Veneto Institute of Molecular Medicine, Padova, Italy.
| |
Collapse
|
4
|
Nitta Y, Osaka J, Maki R, Hakeda-Suzuki S, Suzuki E, Ueki S, Suzuki T, Sugie A. Drosophila model to clarify the pathological significance of OPA1 in autosomal dominant optic atrophy. eLife 2024; 12:RP87880. [PMID: 39177028 PMCID: PMC11343565 DOI: 10.7554/elife.87880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/24/2024] Open
Abstract
Autosomal dominant optic atrophy (DOA) is a progressive form of blindness caused by degeneration of retinal ganglion cells and their axons, mainly caused by mutations in the OPA1 mitochondrial dynamin like GTPase (OPA1) gene. OPA1 encodes a dynamin-like GTPase present in the mitochondrial inner membrane. When associated with OPA1 mutations, DOA can present not only ocular symptoms but also multi-organ symptoms (DOA plus). DOA plus often results from point mutations in the GTPase domain, which are assumed to have dominant-negative effects. However, the presence of mutations in the GTPase domain does not always result in DOA plus. Therefore, an experimental system to distinguish between DOA and DOA plus is needed. In this study, we found that loss-of-function mutations of the dOPA1 gene in Drosophila can imitate the pathology of optic nerve degeneration observed in DOA. We successfully rescued this degeneration by expressing the human OPA1 (hOPA1) gene, indicating that hOPA1 is functionally interchangeable with dOPA1 in the fly system. However, mutations previously identified did not ameliorate the dOPA1 deficiency phenotype. By expressing both WT and DOA plus mutant hOPA1 forms in the optic nerve of dOPA1 mutants, we observed that DOA plus mutations suppressed the rescue, facilitating the distinction between loss-of-function and dominant-negative mutations in hOPA1. This fly model aids in distinguishing DOA from DOA plus and guides initial hOPA1 mutation treatment strategies.
Collapse
Affiliation(s)
- Yohei Nitta
- Brain Research Institute, Niigata UniversityNiigataJapan
| | - Jiro Osaka
- Brain Research Institute, Niigata UniversityNiigataJapan
- School of Life Science and Technology, Tokyo Institute of TechnologyYokohamaJapan
| | - Ryuto Maki
- School of Life Science and Technology, Tokyo Institute of TechnologyYokohamaJapan
| | - Satoko Hakeda-Suzuki
- School of Life Science and Technology, Tokyo Institute of TechnologyYokohamaJapan
- Research Initiatives and Promotion Organization, Yokohama National UniversityYokohamaJapan
| | - Emiko Suzuki
- Department of Biological Sciences, Graduate School of Science, Tokyo Metropolitan UniversityHachiojiJapan
- Department of Gene Function and Phenomics, National Institute of GeneticsMishimaJapan
| | - Satoshi Ueki
- Division of Ophthalmology and Visual Science, Graduate School of Medical and Dental Sciences, Niigata UniversityNiigataJapan
| | - Takashi Suzuki
- School of Life Science and Technology, Tokyo Institute of TechnologyYokohamaJapan
| | - Atsushi Sugie
- Brain Research Institute, Niigata UniversityNiigataJapan
| |
Collapse
|
5
|
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
|
6
|
Lambiri DW, Levin LA. Maculopapillary Bundle Degeneration in Optic Neuropathies. Curr Neurol Neurosci Rep 2024; 24:203-218. [PMID: 38833037 DOI: 10.1007/s11910-024-01343-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/16/2024] [Indexed: 06/06/2024]
Abstract
PURPOSE OF REVIEW Degeneration of the maculopapillary bundle (MPB) is a prominent feature in a spectrum of optic neuropathies. MPB-selective degeneration is seen in specific conditions, such as nutritional and toxic optic neuropathies, Leber hereditary optic neuropathy (LHON), and dominant optic atrophy (DOA). Despite their distinct etiologies and clinical presentations, which encompass variations in age of incidence and monocular or binocular onset, these disorders share a core molecular mechanism: compromised mitochondrial homeostasis. This disruption is characterized by dysfunctions in mitochondrial metabolism, biogenesis, and protein synthesis. This article provides a comprehensive understanding of the MPB's role in optic neuropathies, emphasizing the importance of mitochondrial mechanisms in the pathogenesis of these conditions. RECENT FINDINGS Optical coherence tomography studies have characterized the retinal nerve fiber layer changes accompanying mitochondrial-affiliated optic neuropathies. Selective thinning of the temporal optic nerve head is preceded by thickening in early stages of these disorders which correlates with reductions in macular ganglion cell layer thinning and vascular atrophy. A recently proposed mechanism underpinning the selective atrophy of the MPB involves the positive feedback of reactive oxygen species generation as a common consequence of mitochondrial dysfunction. Additionally, new research has revealed that the MPB can undergo degeneration in the early stages of glaucoma, challenging the historically held belief that this area was not involved in this common optic neuropathy. A variety of anatomical risk factors influence the propensity of glaucomatous MPB degeneration, and cases present distinct patterns of ganglion cell degeneration that are distinct from those observed in mitochondria-associated diseases. This review synthesizes clinical and molecular research on primary MPB disorders, highlighting the commonalities and differences in their pathogenesis. KEY POINTS (BOX) 1. Temporal degeneration of optic nerve fibers accompanied by cecocentral scotoma is a hallmark of maculopapillary bundle (MPB) degeneration. 2. Mechanisms of MPB degeneration commonly implicate mitochondrial dysfunction. 3. Recent research challenges the traditional belief that the MPB is uninvolved in glaucoma by showing degeneration in the early stages of this common optic neuropathy, yet with features distinct from other MPB-selective neuropathies. 4. Reactive oxygen species generation is a mechanism linking mitochondrial mechanisms of MPB-selective optic neuropathies, but in-vivo and in-vitro studies are needed to validate this hypothesis.
Collapse
Affiliation(s)
- Darius W Lambiri
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada
- Department of Ophthalmology and Visual Sciences, McGill University, Montreal, Canada
| | - Leonard A Levin
- Faculty of Medicine and Health Sciences, McGill University, Montreal, Canada.
- Department of Ophthalmology and Visual Sciences, McGill University, Montreal, Canada.
- Department of Neurology & Neurosurgery, McGill University, Montreal, Canada.
| |
Collapse
|
7
|
Borbolis F, Palikaras K. Identifying therapeutic compounds for autosomal dominant optic atrophy (ADOA) through screening in the nematode C. elegans. Methods Cell Biol 2024; 188:89-108. [PMID: 38880530 DOI: 10.1016/bs.mcb.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/18/2024]
Abstract
Autosomal Dominant Optic Atrophy (ADOA) is a rare neurodegenerative condition, characterized by the bilateral loss of vision due to the degeneration of retinal ganglion cells. Its primary cause is linked to mutations in OPA1 gene, which ultimately affect mitochondrial structure and function. The current lack of successful treatments for ADOA emphasizes the need to investigate the mechanisms driving disease pathogenesis and exploit the potential of animal models for preclinical trials. Among such models, Caenorhabditis elegans stands out as a powerful tool, due its simplicity, its genetic tractability, and its relevance to human biology. Despite the lack of a visual system, the presence of mutated OPA1 in the nematode recapitulates ADOA pathology, by stimulating key pathogenic features of the human condition that can be studied in a fast and relatively non-laborious manner. Here, we provide a detailed guide on how to assess the therapeutic efficacy of chemical compounds, in either small or large scale, by evaluating three crucial phenotypes of humanized ADOA model nematodes, that express pathogenic human OPA1 in their GABAergic motor neurons: axonal mitochondria number, neuronal cell death and defecation cycle time. The described methods can deepen our understanding of ADOA pathogenesis and offer a practical framework for developing novel treatment schemes, providing hope for improved therapeutic outcomes and a better quality of life for individuals affected by this currently incurable condition.
Collapse
Affiliation(s)
- Fivos Borbolis
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece; Department of Biology, University of Padova, Padova, Italy
| | - Konstantinos Palikaras
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
8
|
Zanfardino P, Amati A, Doccini S, Cox SN, Tullo A, Longo G, D'Erchia A, Picardi E, Nesti C, Santorelli FM, Petruzzella V. OPA1 mutation affects autophagy and triggers senescence in autosomal dominant optic atrophy plus fibroblasts. Hum Mol Genet 2024; 33:768-786. [PMID: 38280232 DOI: 10.1093/hmg/ddae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 01/06/2024] [Indexed: 01/29/2024] Open
Abstract
In several cases of mitochondrial diseases, the underlying genetic and bioenergetic causes of reduced oxidative phosphorylation (OxPhos) in mitochondrial dysfunction are well understood. However, there is still limited knowledge about the specific cellular outcomes and factors involved for each gene and mutation, which contributes to the lack of effective treatments for these disorders. This study focused on fibroblasts from a patient with Autosomal Dominant Optic Atrophy (ADOA) plus syndrome harboring a mutation in the Optic Atrophy 1 (OPA1) gene. By combining functional and transcriptomic approaches, we investigated the mitochondrial function and identified cellular phenotypes associated with the disease. Our findings revealed that fibroblasts with the OPA1 mutation exhibited a disrupted mitochondrial network and function, leading to altered mitochondrial dynamics and reduced autophagic response. Additionally, we observed a premature senescence phenotype in these cells, suggesting a previously unexplored role of the OPA1 gene in inducing senescence in ADOA plus patients. This study provides novel insights into the mechanisms underlying mitochondrial dysfunction in ADOA plus and highlights the potential importance of senescence in disease progression.
Collapse
Affiliation(s)
- Paola Zanfardino
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of study of Bari Aldo Moro, Piazza G. Cesare, 11, 70124 Bari, Italy
| | - Alessandro Amati
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of study of Bari Aldo Moro, Piazza G. Cesare, 11, 70124 Bari, Italy
| | - Stefano Doccini
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, Viale del Tirreno, 56128 Calambrone, Pisa, Italy
| | - Sharon N Cox
- Department of Biosciences, Biotechnology, and Biopharmaceutics, University of study of Bari Aldo Moro, via Orabona 4, 70125, Bari, Italy
| | - Apollonia Tullo
- Institute of Biomembranes, Bioenergetics and Molecular Biotechnologies (IBIOM), National Research Council, Via G. Amendola 122/O, 70126 Bari, Italy
| | - Giovanna Longo
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of study of Bari Aldo Moro, Piazza G. Cesare, 11, 70124 Bari, Italy
| | - Annamaria D'Erchia
- Department of Biosciences, Biotechnology, and Biopharmaceutics, University of study of Bari Aldo Moro, via Orabona 4, 70125, Bari, Italy
| | - Ernesto Picardi
- Department of Biosciences, Biotechnology, and Biopharmaceutics, University of study of Bari Aldo Moro, via Orabona 4, 70125, Bari, Italy
| | - Claudia Nesti
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, Viale del Tirreno, 56128 Calambrone, Pisa, Italy
| | - Filippo M Santorelli
- Molecular Medicine for Neurodegenerative and Neuromuscular Diseases Unit, IRCCS Stella Maris Foundation, Viale del Tirreno, 56128 Calambrone, Pisa, Italy
| | - Vittoria Petruzzella
- Department of Translational Biomedicine and Neuroscience (DiBraiN), University of study of Bari Aldo Moro, Piazza G. Cesare, 11, 70124 Bari, Italy
| |
Collapse
|
9
|
Wang HL, Siow R, Schmauck-Medina T, Zhang J, Sandset PM, Filshie C, Lund Ø, Partridge L, Bergersen LH, Juel Rasmussen L, Palikaras K, Sotiropoulos I, Storm-Mathisen J, Rubinsztein DC, Spillantini MG, De Zeeuw CI, Watne LO, Vyhnalek M, Veverova K, Liang KX, Tavernarakis N, Bohr VA, Yokote K, Saarela J, Nilsen H, Gonos ES, Scheibye-Knudsen M, Chen G, Kato H, Selbæk G, Fladby T, Nilsson P, Simonsen A, Aarsland D, Lautrup S, Ottersen OP, Cox LS, Fang EF. Meeting Summary of The NYO3 5th NO-Age/AD Meeting and the 1st Norway-UK Joint Meeting on Aging and Dementia: Recent Progress on the Mechanisms and Interventional Strategies. J Gerontol A Biol Sci Med Sci 2024; 79:glae029. [PMID: 38289789 PMCID: PMC10917444 DOI: 10.1093/gerona/glae029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Indexed: 02/01/2024] Open
Abstract
Unhealthy aging poses a global challenge with profound healthcare and socioeconomic implications. Slowing down the aging process offers a promising approach to reduce the burden of a number of age-related diseases, such as dementia, and promoting healthy longevity in the old population. In response to the challenge of the aging population and with a view to the future, Norway and the United Kingdom are fostering collaborations, supported by a "Money Follows Cooperation agreement" between the 2 nations. The inaugural Norway-UK joint meeting on aging and dementia gathered leading experts on aging and dementia from the 2 nations to share their latest discoveries in related fields. Since aging is an international challenge, and to foster collaborations, we also invited leading scholars from 11 additional countries to join this event. This report provides a summary of the conference, highlighting recent progress on molecular aging mechanisms, genetic risk factors, DNA damage and repair, mitophagy, autophagy, as well as progress on a series of clinical trials (eg, using NAD+ precursors). The meeting facilitated dialogue among policymakers, administrative leaders, researchers, and clinical experts, aiming to promote international research collaborations and to translate findings into clinical applications and interventions to advance healthy aging.
Collapse
Affiliation(s)
- He-Ling Wang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Richard Siow
- School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Tomas Schmauck-Medina
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Jianying Zhang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
- Xiangya School of Stomatology, Central South University, Changsha, Hunan, China
| | - Per Morten Sandset
- Department of Haematology, Oslo University Hospital, Oslo, Norway
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | | | | | - Linda Partridge
- Max Planck Institute for Biology of Ageing, Cologne, Germany
- Department of Genetics, Evolution and Environment, Institute of Healthy Ageing, University College London (UCL), London, UK
| | - Linda Hildegard Bergersen
- Brain and Muscle Energy Group, Institute of Oral Biology, University of Oslo, Oslo, Norway
- Center for Healthy Aging, Department of Neuroscience and Pharmacology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Lene Juel Rasmussen
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
| | - Konstantinos Palikaras
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioannis Sotiropoulos
- Institute of Biosciences and Applications NCSR “Demokritos,”Athens, Greece
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, Braga, Portugal
| | - Jon Storm-Mathisen
- Division of Anatomy, Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research, University of Cambridge, Cambridge, UK
- UK Dementia Research Institute, University of Cambridge, Cambridge, UK
| | | | - Chris I De Zeeuw
- Netherlands Institute for Neuroscience, Amsterdam, The Netherlands
- Department of Neuroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Leiv Otto Watne
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
| | - Martin Vyhnalek
- International Clinical Research Centre, St. Anne’s University Hospital, Brno, Czech Republic
- Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | - Katerina Veverova
- Department of Neurology, Second Faculty of Medicine, Charles University and Motol University Hospital, Prague, Czech Republic
| | | | - Nektarios Tavernarakis
- Institute of Molecular Biology and Biotechnology Foundation for Research and Technology, Heraklion, Greece
- Medical School, University of Crete, Heraklion, Greece
| | - Vilhelm A Bohr
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
- Laboratory of Molecular Gerontology, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, USA
| | - Koutaro Yokote
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Janna Saarela
- Centre for Molecular Medicine Norway (NCMM), University of Oslo, Oslo, Norway
- Institute for Molecular Medicine Finland (FIMM), HiLIFE, University of Helsinki, Helsinki, Finland
| | - Hilde Nilsen
- Department of Microbiology, Oslo University Hospital, Oslo, Norway
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| | - Efstathios S Gonos
- National Helenic Research Foundation, Institute of Biology, Medicinal Chemistry and Biotechnology, Athens, Greece
| | - Morten Scheibye-Knudsen
- Department of Cellular and Molecular Medicine, Center for Healthy Aging, University of Copenhagen, Copenhagen, Denmark
- Tracked.bio, Copenhagen, Denmark
| | - Guobing Chen
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou, China
- Department of Microbiology and Immunology, School of Medicine; Institute of Geriatric Immunology, School of Medicine, Jinan University, Guangzhou, China
| | - Hisaya Kato
- Department of Endocrinology, Hematology, and Gerontology, Chiba University Graduate School of Medicine, Chiba, Japan
| | - Geir Selbæk
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Norwegian National Centre for Ageing and Health, Vestfold Hospital Trust, Tønsberg, Norway
| | - Tormod Fladby
- Institute of Clinical Medicine, Campus Ahus, University of Oslo, Oslo, Norway
- Department of Neurology, Akershus University Hospital, Lørenskog, Norway
| | - Per Nilsson
- Department of Neurobiology, Care Sciences and Society, Division of Neurogeriatrics, Center for Alzheimer Research, Karolinska Institutet, Stockholm, Sweden
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital Montebello, Oslo, Norway
| | - Dag Aarsland
- Centre for Age-Related Medicine, Stavanger University Hospital, Stavanger, Norway
- Department of Old Age Psychiatry, Institute of Psychiatry, Psychology, and Neuroscience, King’s College London, London, UK
| | - Sofie Lautrup
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
| | - Ole Petter Ottersen
- Centre for Sustainable Healthcare Education, Faculty of Medicine, University of Oslo, Oslo, Norway
- Karolinska Institutet, Stockholm, Sweden
| | - Lynne S Cox
- Department of Biochemistry, University of Oxford, Oxford, UK
| | - Evandro F Fang
- Department of Clinical Molecular Biology, University of Oslo and Akershus University Hospital, Lørenskog, Norway
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| |
Collapse
|
10
|
Fan C, Wang JX, Xiong ZE, Hu SS, Zhou AJ, Yuan D, Zhang CC, Zhou ZY, Wang T. Saponins from Panax japonicus improve neuronal mitochondrial injury of aging rats. PHARMACEUTICAL BIOLOGY 2023; 61:1401-1412. [PMID: 37667488 PMCID: PMC10484050 DOI: 10.1080/13880209.2023.2244532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 06/16/2023] [Accepted: 07/31/2023] [Indexed: 09/06/2023]
Abstract
CONTEXT Panax japonicus is the dried rhizome of Panax japonicus C.A. Mey. (Araliaceae). Saponins from Panax japonicus (SPJ) exhibit anti-oxidative and anti-aging effects. OBJECTIVE We evaluated the neuroprotective effects of SPJ on aging rats. MATERIALS AND METHODS Sprague-Dawley rats (18-months-old) were randomly divided into aging and SPJ groups (n = 8). Five-month-old rats were taken as the adult control (n = 8). The rats were fed a normal chow diet or the SPJ-containing diet (10 or 30 mg/kg) for 4 months. An in vitro model was established by d-galactose (d-Gal) in the SH-SY5Y cell line and pretreated with SPJ (25 and 50 µg/mL). The neuroprotection of SPJ was evaluated via Nissl staining, flow cytometry, transmission electron microscopy and Western blotting in vivo and in vitro. RESULTS SPJ improved the neuronal degeneration and mitochondrial morphology that are associated with aging. Meanwhile, SPJ up-regulated the protein levels of mitofusin 2 (Mfn2) and optic atrophy 1 (Opa1) and down-regulated the protein level of dynamin-like protein 1 (Drp1) in the hippocampus of aging rats (p < 0.05 or p < 0.01 vs. 22 M). The in vitro studies also demonstrated that SPJ attenuated d-Gal-induced cell senescence concomitant with the improvement in mitochondrial function; SPJ, also up-regulated the Mfn2 and Opa1 protein levels, whereas the Drp1 protein level (p < 0.05 or p < 0.01 vs. d-Gal group) was down-regulated. DISCUSSION AND CONCLUSIONS Further research on the elderly population will contribute to the development and utilization of SPJ for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Cheng Fan
- Institute of Pharmaceutical Process, Academy of Nutrition and Health Hubei Province Key Laboratory of Occupational Hazard Identification and Control School of Medicine, Wuhan University of Science and TechnologyWuhan, China
| | - Jin-Xin Wang
- College of Medical Science, Three Gorges University, Yichang, Hubei, China
| | - Zhang-E. Xiong
- Department of Gastroenterology, Wuhan Third Hospital, Wuhan, China
| | - Shan-Shan Hu
- College of Medical Science, Three Gorges University, Yichang, Hubei, China
| | - Ao-Jia Zhou
- Institute of Pharmaceutical Process, Academy of Nutrition and Health Hubei Province Key Laboratory of Occupational Hazard Identification and Control School of Medicine, Wuhan University of Science and TechnologyWuhan, China
| | - Ding Yuan
- College of Medical Science, Three Gorges University, Yichang, Hubei, China
| | - Chang-Cheng Zhang
- College of Medical Science, Three Gorges University, Yichang, Hubei, China
| | - Zhi-Yong Zhou
- College of Medical Science, Three Gorges University, Yichang, Hubei, China
| | - Ting Wang
- Institute of Pharmaceutical Process, Academy of Nutrition and Health Hubei Province Key Laboratory of Occupational Hazard Identification and Control School of Medicine, Wuhan University of Science and TechnologyWuhan, China
| |
Collapse
|
11
|
Ježek P, Jabůrek M, Holendová B, Engstová H, Dlasková A. Mitochondrial Cristae Morphology Reflecting Metabolism, Superoxide Formation, Redox Homeostasis, and Pathology. Antioxid Redox Signal 2023; 39:635-683. [PMID: 36793196 PMCID: PMC10615093 DOI: 10.1089/ars.2022.0173] [Citation(s) in RCA: 21] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 02/08/2023] [Accepted: 02/09/2023] [Indexed: 02/17/2023]
Abstract
Significance: Mitochondrial (mt) reticulum network in the cell possesses amazing ultramorphology of parallel lamellar cristae, formed by the invaginated inner mitochondrial membrane. Its non-invaginated part, the inner boundary membrane (IBM) forms a cylindrical sandwich with the outer mitochondrial membrane (OMM). Crista membranes (CMs) meet IBM at crista junctions (CJs) of mt cristae organizing system (MICOS) complexes connected to OMM sorting and assembly machinery (SAM). Cristae dimensions, shape, and CJs have characteristic patterns for different metabolic regimes, physiological and pathological situations. Recent Advances: Cristae-shaping proteins were characterized, namely rows of ATP-synthase dimers forming the crista lamella edges, MICOS subunits, optic atrophy 1 (OPA1) isoforms and mitochondrial genome maintenance 1 (MGM1) filaments, prohibitins, and others. Detailed cristae ultramorphology changes were imaged by focused-ion beam/scanning electron microscopy. Dynamics of crista lamellae and mobile CJs were demonstrated by nanoscopy in living cells. With tBID-induced apoptosis a single entirely fused cristae reticulum was observed in a mitochondrial spheroid. Critical Issues: The mobility and composition of MICOS, OPA1, and ATP-synthase dimeric rows regulated by post-translational modifications might be exclusively responsible for cristae morphology changes, but ion fluxes across CM and resulting osmotic forces might be also involved. Inevitably, cristae ultramorphology should reflect also mitochondrial redox homeostasis, but details are unknown. Disordered cristae typically reflect higher superoxide formation. Future Directions: To link redox homeostasis to cristae ultramorphology and define markers, recent progress will help in uncovering mechanisms involved in proton-coupled electron transfer via the respiratory chain and in regulation of cristae architecture, leading to structural determination of superoxide formation sites and cristae ultramorphology changes in diseases. Antioxid. Redox Signal. 39, 635-683.
Collapse
Affiliation(s)
- Petr Ježek
- Department No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Martin Jabůrek
- Department No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Blanka Holendová
- Department No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Hana Engstová
- Department No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| | - Andrea Dlasková
- Department No. 75, Institute of Physiology, Academy of Sciences of the Czech Republic, Prague, Czech Republic
| |
Collapse
|
12
|
Ham SJ, Yoo H, Woo D, Lee DH, Park KS, Chung J. PINK1 and Parkin regulate IP 3R-mediated ER calcium release. Nat Commun 2023; 14:5202. [PMID: 37626046 PMCID: PMC10457342 DOI: 10.1038/s41467-023-40929-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 08/16/2023] [Indexed: 08/27/2023] Open
Abstract
Although defects in intracellular calcium homeostasis are known to play a role in the pathogenesis of Parkinson's disease (PD), the underlying molecular mechanisms remain unclear. Here, we show that loss of PTEN-induced kinase 1 (PINK1) and Parkin leads to dysregulation of inositol 1,4,5-trisphosphate receptor (IP3R) activity, robustly increasing ER calcium release. In addition, we identify that CDGSH iron sulfur domain 1 (CISD1, also known as mitoNEET) functions downstream of Parkin to directly control IP3R. Both genetic and pharmacologic suppression of CISD1 and its Drosophila homolog CISD (also known as Dosmit) restore the increased ER calcium release in PINK1 and Parkin null mammalian cells and flies, respectively, demonstrating the evolutionarily conserved regulatory mechanism of intracellular calcium homeostasis by the PINK1-Parkin pathway. More importantly, suppression of CISD in PINK1 and Parkin null flies rescues PD-related phenotypes including defective locomotor activity and dopaminergic neuronal degeneration. Based on these data, we propose that the regulation of ER calcium release by PINK1 and Parkin through CISD1 and IP3R is a feasible target for treating PD pathogenesis.
Collapse
Affiliation(s)
- Su Jin Ham
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Graduate Program in Genetic Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Heesuk Yoo
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, 08826, Republic of Korea
- Interdisciplinary Graduate Program in Genetic Engineering, Seoul National University, Seoul, 08826, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Daihn Woo
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, 08826, Republic of Korea
| | - Da Hyun Lee
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, 08826, Republic of Korea
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea
| | - Kyu-Sang Park
- Department of Physiology, Yonsei University Wonju College of Medicine, Wonju, 26426, Republic of Korea
- Mitohormesis Research Center, Yonsei University Wonju College of Medicine, Wonju, 26426, Republic of Korea
| | - Jongkyeong Chung
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, 08826, Republic of Korea.
- Interdisciplinary Graduate Program in Genetic Engineering, Seoul National University, Seoul, 08826, Republic of Korea.
- School of Biological Sciences, Seoul National University, Seoul, 08826, Republic of Korea.
| |
Collapse
|
13
|
Ju WK, Perkins GA, Kim KY, Bastola T, Choi WY, Choi SH. Glaucomatous optic neuropathy: Mitochondrial dynamics, dysfunction and protection in retinal ganglion cells. Prog Retin Eye Res 2023; 95:101136. [PMID: 36400670 DOI: 10.1016/j.preteyeres.2022.101136] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 10/04/2022] [Accepted: 11/03/2022] [Indexed: 11/18/2022]
Abstract
Glaucoma is a leading cause of irreversible blindness worldwide and is characterized by a slow, progressive, and multifactorial degeneration of retinal ganglion cells (RGCs) and their axons, resulting in vision loss. Despite its high prevalence in individuals 60 years of age and older, the causing factors contributing to glaucoma progression are currently not well characterized. Intraocular pressure (IOP) is the only proven treatable risk factor. However, lowering IOP is insufficient for preventing disease progression. One of the significant interests in glaucoma pathogenesis is understanding the structural and functional impairment of mitochondria in RGCs and their axons and synapses. Glaucomatous risk factors such as IOP elevation, aging, genetic variation, neuroinflammation, neurotrophic factor deprivation, and vascular dysregulation, are potential inducers for mitochondrial dysfunction in glaucoma. Because oxidative phosphorylation stress-mediated mitochondrial dysfunction is associated with structural and functional impairment of mitochondria in glaucomatous RGCs, understanding the underlying mechanisms and relationship between structural and functional alterations in mitochondria would be beneficial to developing mitochondria-related neuroprotection in RGCs and their axons and synapses against glaucomatous neurodegeneration. Here, we review the current studies focusing on mitochondrial dynamics-based structural and functional alterations in the mitochondria of glaucomatous RGCs and therapeutic strategies to protect RGCs against glaucomatous neurodegeneration.
Collapse
Affiliation(s)
- Won-Kyu Ju
- Hamilton Glaucoma Center and Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA.
| | - Guy A Perkins
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Keun-Young Kim
- National Center for Microscopy and Imaging Research, Department of Neurosciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tonking Bastola
- Hamilton Glaucoma Center and Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA
| | - Woo-Young Choi
- Hamilton Glaucoma Center and Viterbi Family Department of Ophthalmology and Shiley Eye Institute, University of California San Diego, La Jolla, CA, 92093, USA; Department of Plastic Surgery, College of Medicine, Chosun University, Gwang-ju, South Korea
| | - Soo-Ho Choi
- Department of Medicine, University of California San Diego, La Jolla, CA, 92093, USA
| |
Collapse
|
14
|
Liu F, Yuan L, Li L, Yang J, Liu J, Chen Y, Zhang J, Lu Y, Yuan Y, Cheng J. S-sulfhydration of SIRT3 combats BMSC senescence and ameliorates osteoporosis via stabilizing heterochromatic and mitochondrial homeostasis. Pharmacol Res 2023; 192:106788. [PMID: 37146925 DOI: 10.1016/j.phrs.2023.106788] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 04/23/2023] [Accepted: 05/02/2023] [Indexed: 05/07/2023]
Abstract
Senescence of bone marrow mesenchymal stem cells (BMSCs) is one of the leading causes of osteoporosis. SIRT3, an essential NAD-dependent histone deacetylase, is highly correlated with BMSC senescence-mediated bone degradation and mitochondrial/heterochromatic disturbance. S-sulfhydration of cysteine residues favorably enhances SIRT3 activity by forming persulfides. Nevertheless, the underlying molecular mechanism of SIRT3 S-sulfhydration on mitochondrial/heterochromatic homeostasis involved in BMSC senescence remains unknown. Here, we demonstrated that CBS and CSE, endogenous hydrogen sulfide synthases, are downregulated with BMSC senescence. Exogenous H2S donor NaHS-mediated SIRT3 augmentation rescued the senescent phenotypes of BMSCs. Conversely, SIRT3 deletion accelerated oxidative stress-induced BMSC senescence through mitochondrial dysfunction and the detachment of the heterochromatic protein H3K9me3 from the nuclear envelope protein Lamin B1. H2S-mediated SIRT3 S-sulfhydration modification rescued the disorganized heterochromatin and fragmented mitochondria induced by the S-sulfhydration inhibitor dithiothreitol, thus leading to elevated osteogenic capacity and preventing BMSC senescence. The antisenescence effect of S-sulfhydration modification on BMSCs was abolished when the CXXC sites of the SIRT3 zinc finger motif were mutated. In vivo, aged mice-derived BMSCs pretreated with NaHS were orthotopically transplanted to the ovariectomy-induced osteoporotic mice, and we proved that SIRT3 ameliorates bone loss by inhibiting BMSC senescence. Overall, our study for the first time indicates a novel role of SIRT3 S-sulfhydration in stabilizing heterochromatin and mitochondrial homeostasis in counteracting BMSC senescence, providing a potential target for the treatment of degenerative bone diseases.
Collapse
Affiliation(s)
- Fei Liu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Longhui Yuan
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lan Li
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingchao Yang
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jingping Liu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Younan Chen
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Jie Zhang
- Core Facility of West China Hospital, Sichuan University, Chengdu P.R. China
| | - Yanrong Lu
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yujia Yuan
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Jingqiu Cheng
- Department of Endocrinology & Metabolism and Key Lab of Transplant Engineering and Immunology, NHFPC, West China Hospital, Sichuan University, Chengdu 610041, China; Department of Endocrinology & Metabolism and Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
15
|
Mitochondrial Lon-induced mitophagy benefits hypoxic resistance via Ca 2+-dependent FUNDC1 phosphorylation at the ER-mitochondria interface. Cell Death Dis 2023; 14:199. [PMID: 36927870 PMCID: PMC10020552 DOI: 10.1038/s41419-023-05723-1] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 02/18/2023] [Accepted: 03/03/2023] [Indexed: 03/18/2023]
Abstract
During hypoxia, FUNDC1 acts as a mitophagy receptor and accumulates at the ER (endoplasmic reticulum)-mitochondria contact sites (EMC), also called mitochondria-associated membranes (MAM). In mitophagy, the ULK1 complex phosphorylates FUNDC1(S17) at the EMC site. However, how mitochondria sense the stress and send the signal from the inside to the outside of mitochondria to trigger mitophagy is still unclear. Mitochondrial Lon was reported to be localized at the EMC under stress although the function remained unknown. In this study, we explored the mechanism of how mitochondrial sensors of hypoxia trigger and stabilize the FUNDC1-ULK1 complex by Lon in the EMC for cell survival and cancer progression. We demonstrated that Lon is accumulated in the EMC and associated with FUNDC1-ULK1 complex to induce mitophagy via chaperone activity under hypoxia. Intriguingly, we found that Lon-induced mitophagy is through binding with mitochondrial Na+/Ca2+ exchanger (NCLX) to promote FUNDC1-ULK1-mediated mitophagy at the EMC site in vitro and in vivo. Accordingly, our findings highlight a novel mechanism responsible for mitophagy initiation under hypoxia by chaperone Lon in mitochondria through the interaction with FUNDC1-ULK1 complex at the EMC site. These findings provide a direct correlation between Lon and mitophagy on cell survival and cancer progression.
Collapse
|
16
|
Caenorhabditis elegans as a Model System to Study Human Neurodegenerative Disorders. Biomolecules 2023; 13:biom13030478. [PMID: 36979413 PMCID: PMC10046667 DOI: 10.3390/biom13030478] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 02/18/2023] [Accepted: 03/01/2023] [Indexed: 03/08/2023] Open
Abstract
In recent years, advances in science and technology have improved our quality of life, enabling us to tackle diseases and increase human life expectancy. However, longevity is accompanied by an accretion in the frequency of age-related neurodegenerative diseases, creating a growing burden, with pervasive social impact for human societies. The cost of managing such chronic disorders and the lack of effective treatments highlight the need to decipher their molecular and genetic underpinnings, in order to discover new therapeutic targets. In this effort, the nematode Caenorhabditis elegans serves as a powerful tool to recapitulate several disease-related phenotypes and provides a highly malleable genetic model that allows the implementation of multidisciplinary approaches, in addition to large-scale genetic and pharmacological screens. Its anatomical transparency allows the use of co-expressed fluorescent proteins to track the progress of neurodegeneration. Moreover, the functional conservation of neuronal processes, along with the high homology between nematode and human genomes, render C. elegans extremely suitable for the study of human neurodegenerative disorders. This review describes nematode models used to study neurodegeneration and underscores their contribution in the effort to dissect the molecular basis of human diseases and identify novel gene targets with therapeutic potential.
Collapse
|
17
|
Sánchez-Rodríguez R, Tezze C, Agnellini AHR, Angioni R, Venegas FC, Cioccarelli C, Munari F, Bertoldi N, Canton M, Desbats MA, Salviati L, Gissi R, Castegna A, Soriano ME, Sandri M, Scorrano L, Viola A, Molon B. OPA1 drives macrophage metabolism and functional commitment via p65 signaling. Cell Death Differ 2023; 30:742-752. [PMID: 36307526 PMCID: PMC9984365 DOI: 10.1038/s41418-022-01076-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 09/26/2022] [Accepted: 10/06/2022] [Indexed: 11/05/2022] Open
Abstract
Macrophages are essential players for the host response against pathogens, regulation of inflammation and tissue regeneration. The wide range of macrophage functions rely on their heterogeneity and plasticity that enable a dynamic adaptation of their responses according to the surrounding environmental cues. Recent studies suggest that metabolism provides synergistic support for macrophage activation and elicitation of desirable immune responses; however, the metabolic pathways orchestrating macrophage activation are still under scrutiny. Optic atrophy 1 (OPA1) is a mitochondria-shaping protein controlling mitochondrial fusion, cristae biogenesis and respiration; clear evidence shows that the lack or dysfunctional activity of this protein triggers the accumulation of metabolic intermediates of the TCA cycle. In this study, we show that OPA1 has a crucial role in macrophage activation. Selective Opa1 deletion in myeloid cells impairs M1-macrophage commitment. Mechanistically, Opa1 deletion leads to TCA cycle metabolite accumulation and defective NF-κB signaling activation. In an in vivo model of muscle regeneration upon injury, Opa1 knockout macrophages persist within the damaged tissue, leading to excess collagen deposition and impairment in muscle regeneration. Collectively, our data indicate that OPA1 is a key metabolic driver of macrophage functions.
Collapse
Affiliation(s)
- Ricardo Sánchez-Rodríguez
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy
| | - Caterina Tezze
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine, 35129, Padova, Italy
| | | | - Roberta Angioni
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy
| | - Francisca C Venegas
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy
| | - Chiara Cioccarelli
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy
| | - Fabio Munari
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy
| | - Nicole Bertoldi
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy
| | - Marcella Canton
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy
| | - Maria Andrea Desbats
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Leonardo Salviati
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy
- Clinical Genetics Unit, Department of Women's and Children's Health, University of Padova, Padova, Italy
| | - Rosanna Gissi
- Department of Biosciences, Biotechnologies and Environment, 70125, Bari, Italy
| | - Alessandra Castegna
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy
- Department of Biosciences, Biotechnologies and Environment, 70125, Bari, Italy
| | | | - Marco Sandri
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy
- Veneto Institute of Molecular Medicine, 35129, Padova, Italy
- Department of Medicine, McGill University, Montreal, Montreal (Quebec), H4A 3J1, Canada
| | - Luca Scorrano
- Department of Biology, University of Padova, 35131, Padova, Italy
| | - Antonella Viola
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy.
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy.
| | - Barbara Molon
- Department of Biomedical Sciences, University of Padova, 35131, Padova, Italy.
- Istituto di Ricerca Pediatrica IRP- Fondazione Città della Speranza, 35127, Padova, Italy.
| |
Collapse
|
18
|
Characterization of Primary Cilia Formation in Human ESC-Derived Retinal Organoids. Stem Cells Int 2023; 2023:6494486. [PMID: 36684387 PMCID: PMC9859708 DOI: 10.1155/2023/6494486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 12/07/2022] [Accepted: 12/31/2022] [Indexed: 01/15/2023] Open
Abstract
Objectives Primary cilia are conserved organelles found in polarized mammalian cells that regulate neuronal growth, migration, and differentiation. Proper cilia formation is essential during eye development. Our previous reports found that both amacrine and retinal ganglion cells (RGCs) contain primary cilia in primate and rodent retinas. However, whether primary cilia are present in the inner retina of human retinal organoids remains unknown. The purpose of this study is to characterize the primary cilia distribution in human embryonic stem cell (hESC-derived retinal organoid development. Materials and Methods Retinal organoids were differentiated from a hESC line, harvested at various developmental timepoints (day 44-day 266), and immunostained with antibodies for primary cilia, including Arl13b (for the axoneme), AC3, and Centrin3 (for the basal body). AP2α, Prox1, GAD67, Calretinin, GFAP, PKCα, and Chx10 antibodies as well as Brn3b-promoted tdTomato expression were used to visualize retinal cell types. Results A group of ciliated cells were present in the inner aspects of retinal organoids from day 44 to day 266 in culture. Ciliated Chx10-positive retinal progenitor cells, GFAP-positive astrocytes, and PKCα-positive rod-bipolar cells were detected later during development (day 176 to day 266). Ciliation persisted during all stages of retinal developmental in AP2α-positive amacrine cells, but it was decreased in Brn3b-positive retinal ganglion cells (RGCs) at later time points. Additionally, AC3-positive astrocytes significantly decreased during the later stages of organoid formation. Conclusions Amacrine cells in retinal organoids retain cilia throughout development, whereas RGC ciliation gradually and progressively decreases with organoid maturation.
Collapse
|
19
|
Charmpilas N, Fang EF, Palikaras K. Mitophagy and Neuroinflammation: A Compelling Interplay. Curr Neuropharmacol 2023; 21:1477-1481. [PMID: 35762540 PMCID: PMC10472808 DOI: 10.2174/1570159x20666220628153632] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Revised: 04/29/2022] [Accepted: 06/23/2022] [Indexed: 11/22/2022] Open
Abstract
Mitochondria are the main sites of energy production and a major source of metabolic stress. Not surprisingly, impairment of mitochondrial homeostasis is strongly associated with the development and progression of a broad spectrum of human pathologies, including neurodegenerative disorders. Mitophagy mediates the selective degradation of damaged organelles, thus promoting cellular viability and tissue integrity. Defective mitophagy triggers cellular senescence and prolonged neuroinflammation, leading eventually to cell death and brain homeostasis collapse. Here, we survey the intricate interplay between mitophagy and neuroinflammation, highlighting that mitophagy can be a focal point for therapeutic interventions to tackle neurodegeneration.
Collapse
Affiliation(s)
- Nikolaos Charmpilas
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Ageing-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Evandro Fei Fang
- Department of Clinical Molecular Biology, University of Oslo, Oslo, Norway
- Akershus University Hospital, Lørenskog, Norway
- The Norwegian Centre on Healthy Ageing (NO-Age), Oslo, Norway
| | - Konstantinos Palikaras
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
20
|
Sánchez-González C, Herrero Martín JC, Salegi Ansa B, Núñez de Arenas C, Stančič B, Pereira MP, Contreras L, Cuezva JM, Formentini L. Chronic inhibition of the mitochondrial ATP synthase in skeletal muscle triggers sarcoplasmic reticulum distress and tubular aggregates. Cell Death Dis 2022; 13:561. [PMID: 35732639 PMCID: PMC9217934 DOI: 10.1038/s41419-022-05016-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2021] [Revised: 06/03/2022] [Accepted: 06/09/2022] [Indexed: 01/21/2023]
Abstract
Tubular aggregates (TA) are honeycomb-like arrays of sarcoplasmic-reticulum (SR) tubules affecting aged glycolytic fibers of male individuals and inducing severe sarcomere disorganization and muscular pain. TA develop in skeletal muscle from Tubular Aggregate Myopathy (TAM) patients as well as in other disorders including endocrine syndromes, diabetes, and ageing, being their primary cause unknown. Nowadays, there is no cure for TA. Intriguingly, both hypoxia and calcium dyshomeostasis prompt TA formation, pointing to a possible role for mitochondria in their setting. However, a functional link between mitochondrial dysfunctions and TA remains unknown. Herein, we investigate the alteration in muscle-proteome of TAM patients, the molecular mechanism of TA onset and a potential therapy in a preclinical mouse model of the disease. We show that in vivo chronic inhibition of the mitochondrial ATP synthase in muscle causes TA. Upon long-term restrained oxidative phosphorylation (OXPHOS), oxidative soleus experiments a metabolic and structural switch towards glycolytic fibers, increases mitochondrial fission, and activates mitophagy to recycle damaged mitochondria. TA result from the overresponse of the fission controller DRP1, that upregulates the Store-Operate-Calcium-Entry and increases the mitochondria-SR interaction in a futile attempt to buffer calcium overloads upon prolonged OXPHOS inhibition. Accordingly, hypoxic muscles cultured ex vivo show an increase in mitochondria/SR contact sites and autophagic/mitophagic zones, where TA clusters grow around defective mitochondria. Moreover, hypoxia triggered a stronger TA formation upon ATP synthase inhibition, and this effect was reduced by the DRP1 inhibitor mDIVI. Remarkably, the muscle proteome of TAM patients displays similar alterations in mitochondrial dynamics and in ATP synthase contents. In vivo edaravone treatment in mice with restrained OXPHOS restored a healthy phenotype by prompting mitogenesis and mitochondrial fusion. Altogether, our data provide a functional link between the ATP synthase/DRP1 axis and the setting of TA, and repurpose edaravone as a possible treatment for TA-associated disorders.
Collapse
Affiliation(s)
- Cristina Sánchez-González
- grid.5515.40000000119578126Departamento de Biología Molecular, Centro de Biología Molecular ‘“Severo Ochoa’” (CBMSO), c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan Cruz Herrero Martín
- grid.5515.40000000119578126Departamento de Biología Molecular, Centro de Biología Molecular ‘“Severo Ochoa’” (CBMSO), c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Beñat Salegi Ansa
- grid.5515.40000000119578126Departamento de Biología Molecular, Centro de Biología Molecular ‘“Severo Ochoa’” (CBMSO), c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Núñez de Arenas
- grid.5515.40000000119578126Departamento de Biología Molecular, Centro de Biología Molecular ‘“Severo Ochoa’” (CBMSO), c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
| | - Brina Stančič
- grid.5515.40000000119578126Departamento de Biología Molecular, Centro de Biología Molecular ‘“Severo Ochoa’” (CBMSO), c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain
| | - Marta P. Pereira
- grid.5515.40000000119578126Departamento de Biología Molecular, Centro de Biología Molecular ‘“Severo Ochoa’” (CBMSO), c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain ,grid.5515.40000000119578126Instituto Universitario de Biología Molecular, IUBM, Universidad Autónoma de Madrid, Madrid, Spain
| | - Laura Contreras
- grid.5515.40000000119578126Departamento de Biología Molecular, Centro de Biología Molecular ‘“Severo Ochoa’” (CBMSO), c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain ,grid.5515.40000000119578126Instituto Universitario de Biología Molecular, IUBM, Universidad Autónoma de Madrid, Madrid, Spain ,grid.419651.e0000 0000 9538 1950Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD), Madrid, Spain
| | - José M. Cuezva
- grid.5515.40000000119578126Departamento de Biología Molecular, Centro de Biología Molecular ‘“Severo Ochoa’” (CBMSO), c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain ,grid.5515.40000000119578126Instituto Universitario de Biología Molecular, IUBM, Universidad Autónoma de Madrid, Madrid, Spain ,grid.512044.60000 0004 7666 5367Instituto de Investigación Hospital 12 de Octubre, i+12, Madrid, Spain
| | - Laura Formentini
- grid.5515.40000000119578126Departamento de Biología Molecular, Centro de Biología Molecular ‘“Severo Ochoa’” (CBMSO), c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, Madrid, Spain ,grid.452372.50000 0004 1791 1185Centro de Investigación Biomédica en red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain ,grid.5515.40000000119578126Instituto Universitario de Biología Molecular, IUBM, Universidad Autónoma de Madrid, Madrid, Spain ,grid.512044.60000 0004 7666 5367Instituto de Investigación Hospital 12 de Octubre, i+12, Madrid, Spain
| |
Collapse
|
21
|
Cheng XT, Huang N, Sheng ZH. Programming axonal mitochondrial maintenance and bioenergetics in neurodegeneration and regeneration. Neuron 2022; 110:1899-1923. [PMID: 35429433 PMCID: PMC9233091 DOI: 10.1016/j.neuron.2022.03.015] [Citation(s) in RCA: 77] [Impact Index Per Article: 38.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/04/2022] [Accepted: 03/10/2022] [Indexed: 12/11/2022]
Abstract
Mitochondria generate ATP essential for neuronal growth, function, and regeneration. Due to their polarized structures, neurons face exceptional challenges to deliver mitochondria to and maintain energy homeostasis throughout long axons and terminal branches where energy is in high demand. Chronic mitochondrial dysfunction accompanied by bioenergetic failure is a pathological hallmark of major neurodegenerative diseases. Brain injury triggers acute mitochondrial damage and a local energy crisis that accelerates neuron death. Thus, mitochondrial maintenance defects and axonal energy deficits emerge as central problems in neurodegenerative disorders and brain injury. Recent studies have started to uncover the intrinsic mechanisms that neurons adopt to maintain (or reprogram) axonal mitochondrial density and integrity, and their bioenergetic capacity, upon sensing energy stress. In this review, we discuss recent advances in how neurons maintain a healthy pool of axonal mitochondria, as well as potential therapeutic strategies that target bioenergetic restoration to power neuronal survival, function, and regeneration.
Collapse
Affiliation(s)
- Xiu-Tang Cheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Ning Huang
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA
| | - Zu-Hang Sheng
- Synaptic Function Section, The Porter Neuroscience Research Center, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Room 2B-215, 35 Convent Drive, Bethesda, MD 20892-3706, USA.
| |
Collapse
|
22
|
Boccuni I, Fairless R. Retinal Glutamate Neurotransmission: From Physiology to Pathophysiological Mechanisms of Retinal Ganglion Cell Degeneration. Life (Basel) 2022; 12:638. [PMID: 35629305 PMCID: PMC9147752 DOI: 10.3390/life12050638] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 04/22/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
Glutamate neurotransmission and metabolism are finely modulated by the retinal network, where the efficient processing of visual information is shaped by the differential distribution and composition of glutamate receptors and transporters. However, disturbances in glutamate homeostasis can result in glutamate excitotoxicity, a major initiating factor of common neurodegenerative diseases. Within the retina, glutamate excitotoxicity can impair visual transmission by initiating degeneration of neuronal populations, including retinal ganglion cells (RGCs). The vulnerability of RGCs is observed not just as a result of retinal diseases but has also been ascribed to other common neurodegenerative and peripheral diseases. In this review, we describe the vulnerability of RGCs to glutamate excitotoxicity and the contribution of different glutamate receptors and transporters to this. In particular, we focus on the N-methyl-d-aspartate (NMDA) receptor as the major effector of glutamate-induced mechanisms of neurodegeneration, including impairment of calcium homeostasis, changes in gene expression and signalling, and mitochondrial dysfunction, as well as the role of endoplasmic reticular stress. Due to recent developments in the search for modulators of NMDA receptor signalling, novel neuroprotective strategies may be on the horizon.
Collapse
Affiliation(s)
- Isabella Boccuni
- Institute for Physiology and Pathophysiology, Heidelberg University, 69120 Heidelberg, Germany
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
| | - Richard Fairless
- Department of Neurology, University Clinic Heidelberg, 69120 Heidelberg, Germany;
- Clinical Cooperation Unit (CCU) Neurooncology, German Cancer Consortium (DKTK), German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
23
|
VDAC1 regulates neuronal cell loss after retinal trauma injury by a mitochondria-independent pathway. Cell Death Dis 2022; 13:393. [PMID: 35449127 PMCID: PMC9023530 DOI: 10.1038/s41419-022-04755-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 03/04/2022] [Accepted: 03/16/2022] [Indexed: 12/23/2022]
Abstract
The voltage-dependent anion channel 1 (VDAC1) was first described as a mitochondrial porin that mediates the flux of metabolites and ions, thereby integrating both cell survival and death signals. In the nervous system, the functional roles of VDAC1 remain poorly understood. Herein, the rat retina was employed to study VDAC1. First, it was observed that even subtle changes in VDAC1 levels affect neuronal survival, inducing severe alterations in the retinal morphology. We next examined the regulation of VDAC1 after traumatic retinal injury. After mechanical trauma, SOD1 translocates towards the nucleus, which is insufficient to contain the consequences of oxidative stress, as determined by the evaluation of protein carbonylation. Using in vitro models of oxidative stress and mechanical injury in primary retinal cell cultures, it was possible to determine that inhibition of VDAC1 oligomerization by 4'-diisothiocyano-2,2'-disulfonic acid stilbene (DIDS) rescues cell viability, impacting microglial cell activation. We next focused on the regulation of VDAC1 after retinal mechanical injury. VDAC1 was promptly upregulated 2 h after lesion in the plasma membrane and endoplasmic reticulum rather than in the mitochondria, and multimers of VDAC1 were assembled after lesion. DIDS intraocular application decreased apoptosis and prevented microglial polarization, which confirmed in vitro observations. Considering the role of microglia in neuroinflammation, multiplex evaluation of cytokines showed that DIDS application disorganized the inflammatory response 2 h after the lesion, matching the fast regulation of VDAC1. Taken together, data disclosed that fine regulation of VDAC1 influences neuronal survival, and pharmacological inhibition after trauma injury has neuroprotective effects. This protection may be attributed to the effects on VDAC1 abnormal accumulation in the plasma membrane, thereby controlling the activation of microglial cells. We concluded that VDAC1 is a putative therapeutic target in neuronal disorders since it integrates both death and survival cellular signaling.
Collapse
|
24
|
Cartes-Saavedra B, Macuada J, Lagos D, Arancibia D, Andrés ME, Yu-Wai-Man P, Hajnóczky G, Eisner V. OPA1 Modulates Mitochondrial Ca 2+ Uptake Through ER-Mitochondria Coupling. Front Cell Dev Biol 2022; 9:774108. [PMID: 35047497 PMCID: PMC8762365 DOI: 10.3389/fcell.2021.774108] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2021] [Accepted: 11/30/2021] [Indexed: 11/16/2022] Open
Abstract
Autosomal Dominant Optic Atrophy (ADOA), a disease that causes blindness and other neurological disorders, is linked to OPA1 mutations. OPA1, dependent on its GTPase and GED domains, governs inner mitochondrial membrane (IMM) fusion and cristae organization, which are central to oxidative metabolism. Mitochondrial dynamics and IMM organization have also been implicated in Ca2+ homeostasis and signaling but the specific involvements of OPA1 in Ca2+ dynamics remain to be established. Here we studied the possible outcomes of OPA1 and its ADOA-linked mutations in Ca2+ homeostasis using rescue and overexpression strategies in Opa1-deficient and wild-type murine embryonic fibroblasts (MEFs), respectively and in human ADOA-derived fibroblasts. MEFs lacking Opa1 required less Ca2+ mobilization from the endoplasmic reticulum (ER) to induce a mitochondrial matrix [Ca2+] rise ([Ca2+]mito). This was associated with closer ER-mitochondria contacts and no significant changes in the mitochondrial calcium uniporter complex. Patient cells carrying OPA1 GTPase or GED domain mutations also exhibited altered Ca2+ homeostasis, and the mutations associated with lower OPA1 levels displayed closer ER-mitochondria gaps. Furthermore, in Opa1 -/- MEF background, we found that acute expression of OPA1 GTPase mutants but no GED mutants, partially restored cytosolic [Ca2+] ([Ca2+]cyto) needed for a prompt [Ca2+]mito rise. Finally, OPA1 mutants' overexpression in WT MEFs disrupted Ca2+ homeostasis, partially recapitulating the observations in ADOA patient cells. Thus, OPA1 modulates functional ER-mitochondria coupling likely through the OPA1 GED domain in Opa1 -/- MEFs. However, the co-existence of WT and mutant forms of OPA1 in patients promotes an imbalance of Ca2+ homeostasis without a domain-specific effect, likely contributing to the overall ADOA progress.
Collapse
Affiliation(s)
- Benjamín Cartes-Saavedra
- Departamento Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Josefa Macuada
- Departamento Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Daniel Lagos
- Departamento Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Duxan Arancibia
- Departamento Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - María E. Andrés
- Departamento Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Patrick Yu-Wai-Man
- UCL Institute of Ophthalmology, University College London, London, United Kingdom
- Moorfields Eye Hospital NHS Foundation Trust, London, United Kingdom
- Cambridge Eye Unit, Addenbrooke’s Hospital, Cambridge University Hospitals, Cambridge, United Kingdom
- John van Geest Centre for Brain Repair and MRC Mitochondrial Biology Unit, Department of Clinical Neurosciences, University of Cambridge, Cambridge, United Kingdom
| | - György Hajnóczky
- MitoCare Center for Mitochondrial Imaging Research and Diagnostics, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Verónica Eisner
- Departamento Biología Celular y Molecular, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|