1
|
Abokyi S, Tse DYY. Age-related driving mechanisms of retinal diseases and neuroprotection by transcription factor EB-targeted therapy. Neural Regen Res 2025; 20:366-377. [PMID: 38819040 PMCID: PMC11317960 DOI: 10.4103/nrr.nrr-d-23-02033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Revised: 02/08/2024] [Accepted: 03/07/2024] [Indexed: 06/01/2024] Open
Abstract
Retinal aging has been recognized as a significant risk factor for various retinal disorders, including diabetic retinopathy, age-related macular degeneration, and glaucoma, following a growing understanding of the molecular underpinnings of their development. This comprehensive review explores the mechanisms of retinal aging and investigates potential neuroprotective approaches, focusing on the activation of transcription factor EB. Recent meta-analyses have demonstrated promising outcomes of transcription factor EB-targeted strategies, such as exercise, calorie restriction, rapamycin, and metformin, in patients and animal models of these common retinal diseases. The review critically assesses the role of transcription factor EB in retinal biology during aging, its neuroprotective effects, and its therapeutic potential for retinal disorders. The impact of transcription factor EB on retinal aging is cell-specific, influencing metabolic reprogramming and energy homeostasis in retinal neurons through the regulation of mitochondrial quality control and nutrient-sensing pathways. In vascular endothelial cells, transcription factor EB controls important processes, including endothelial cell proliferation, endothelial tube formation, and nitric oxide levels, thereby influencing the inner blood-retinal barrier, angiogenesis, and retinal microvasculature. Additionally, transcription factor EB affects vascular smooth muscle cells, inhibiting vascular calcification and atherogenesis. In retinal pigment epithelial cells, transcription factor EB modulates functions such as autophagy, lysosomal dynamics, and clearance of the aging pigment lipofuscin, thereby promoting photoreceptor survival and regulating vascular endothelial growth factor A expression involved in neovascularization. These cell-specific functions of transcription factor EB significantly impact retinal aging mechanisms encompassing proteostasis, neuronal synapse plasticity, energy metabolism, microvasculature, and inflammation, ultimately offering protection against retinal aging and diseases. The review emphasizes transcription factor EB as a potential therapeutic target for retinal diseases. Therefore, it is imperative to obtain well-controlled direct experimental evidence to confirm the efficacy of transcription factor EB modulation in retinal diseases while minimizing its risk of adverse effects.
Collapse
Affiliation(s)
- Samuel Abokyi
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
- Research Center for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
| | - Dennis Yan-yin Tse
- School of Optometry, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
- Research Center for SHARP Vision, The Hong Kong Polytechnic University, Kowloon, Hong Kong Special Administrative Region, China
- Center for Eye and Vision Research, Sha Tin, Hong Kong Special Administrative Region, China
| |
Collapse
|
2
|
Morgan JE, Bevan RJ, Cimaglia G. Retinal Ganglion Cell Subtypes and Their Vulnerability in Glaucoma. Methods Mol Biol 2025; 2858:191-205. [PMID: 39433677 DOI: 10.1007/978-1-0716-4140-8_16] [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: 10/23/2024]
Abstract
The detection of selective retinal ganglion cell damage in glaucoma has been a long sought-after goal, not just for the development of clinical tests for the early detection of glaucoma but for the elucidation of potential mechanisms underlying retinal ganglion cell loss. Early reports of the selective vulnerability of larger retinal ganglion cells (RGCs) in human studies did not translate simply to the loss of a particular class of RGC but more likely reflected shrinkage and degeneration across all RGC classes. Subsequent studies of nonhuman primate (NHP) models of glaucoma indicated some selectivity with great damage to the magnocellular vs parvocellular pathways. More recently, rodent models of experimental glaucoma have highlighted a selective vulnerability of OFF-centered RGCs-particularly those with transient responses. Selectivity for OFF pathway damage is also seen as a trend in a rat model of glaucoma. These data support the concept that some RGCs are more vulnerable to the effects of glaucoma damage. This chapter covers some of the methods to elucidate RGC damage and the relevance of model selection to mimic human glaucoma rather than just RGC death.
Collapse
Affiliation(s)
- James E Morgan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK.
| | - Ryan J Bevan
- UK Dementia Research Institute, Hadyn Ellis Building, Cardiff University, Cardiff, UK
| | - Gloria Cimaglia
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, UK
- UK Dementia Research Institute, Hadyn Ellis Building, Cardiff University, Cardiff, UK
| |
Collapse
|
3
|
Otmani A, Jóhannesson G, Brautaset R, Tribble JR, Williams PA. Prophylactic nicotinamide treatment protects from rotenone-induced neurodegeneration by increasing mitochondrial content and volume. Acta Neuropathol Commun 2024; 12:37. [PMID: 38429841 PMCID: PMC10908050 DOI: 10.1186/s40478-024-01724-z] [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: 11/27/2023] [Accepted: 01/02/2024] [Indexed: 03/03/2024] Open
Abstract
Leber's hereditary optic neuropathy (LHON) is driven by mtDNA mutations affecting Complex I presenting as progressive retinal ganglion cell dysfunction usually in the absence of extra-ophthalmic symptoms. There are no long-term neuroprotective agents for LHON. Oral nicotinamide provides a robust neuroprotective effect against mitochondrial and metabolic dysfunction in other retinal injuries. We explored the potential for nicotinamide to protect mitochondria in LHON by modelling the disease in mice through intravitreal injection of the Complex I inhibitor rotenone. Using MitoV mice expressing a mitochondrial-tagged YFP in retinal ganglion cells we assessed mitochondrial morphology through super-resolution imaging and digital reconstruction. Rotenone induced Complex I inhibition resulted in retinal ganglion cell wide mitochondrial loss and fragmentation. This was prevented by oral nicotinamide treatment. Mitochondrial ultrastructure was quantified by transition electron microscopy, demonstrating a loss of cristae density following rotenone injection, which was also prevented by nicotinamide treatment. These results demonstrate that nicotinamide protects mitochondria during Complex I dysfunction. Nicotinamide has the potential to be a useful treatment strategy for LHON to limit retinal ganglion cell degeneration.
Collapse
Affiliation(s)
- Amin Otmani
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64, Stockholm, Sweden
| | - Gauti Jóhannesson
- Department of Clinical Sciences, Ophthalmology, Umeå University, Umeå, Sweden
- Wallenberg Centre of Molecular Medicine, Umeå University, Umeå, Sweden
- Department of Ophthalmology, University of Iceland, Reykjavik, Iceland
| | - Rune Brautaset
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64, Stockholm, Sweden
| | - James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64, Stockholm, Sweden.
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 171 64, Stockholm, Sweden.
| |
Collapse
|
4
|
Atamena D, Gurram V, Petsophonsakul P, Khosrobakhsh F, Arrázola MS, Botella M, Wissinger B, Szelechowski M, Belenguer P. Genetic background modulates phenotypic expressivity in OPA1 mutated mice, relevance to DOA pathogenesis. Front Mol Neurosci 2023; 16:1241222. [PMID: 37736113 PMCID: PMC10510408 DOI: 10.3389/fnmol.2023.1241222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/14/2023] [Indexed: 09/23/2023] Open
Abstract
Dominant optic atrophy (DOA) is mainly caused by OPA1 mutations and is characterized by the degeneration of retinal ganglion cells (RGCs), whose axons form the optic nerve. The penetrance of DOA is incomplete and the disease is marked by highly variable expressivity, ranging from asymptomatic patients to some who are totally blind or who suffer from multisystemic effects. No clear genotype-phenotype correlation has been established to date. Taken together, these observations point toward the existence of modifying genetic and/or environmental factors that modulate disease severity. Here, we investigated the influence of genetic background on DOA expressivity by switching the previously described DOA mouse model bearing the c.1065 + 5G → A Opa1 mutation from mixed C3H; C57BL/6 J to a pure C57BL/6 J background. We no longer observed retinal and optic nerve abnormalities; the findings indicated no degeneration, but rather a sex-dependent negative effect on RGC connectivity. This highlights the fact that RGC synaptic alteration might precede neuronal death, as has been proposed in other neurodegenerative diseases, providing new clinical considerations for early diagnosis as well as a new therapeutic window for DOA. Furthermore, our results demonstrate the importance of secondary genetic factors in the variability of DOA expressivity and offer a model for screening for aggravating environmental and genetic factors.
Collapse
Affiliation(s)
- Djamaa Atamena
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université Toulouse III, CNRS, Toulouse, France
| | - Venu Gurram
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université Toulouse III, CNRS, Toulouse, France
| | - Petnoï Petsophonsakul
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université Toulouse III, CNRS, Toulouse, France
| | - Farnoosh Khosrobakhsh
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université Toulouse III, CNRS, Toulouse, France
- Department of Biological Science, University of Kurdistan, Sanandaj, Iran
| | - Macarena S. Arrázola
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université Toulouse III, CNRS, Toulouse, France
| | - Marlène Botella
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université Toulouse III, CNRS, Toulouse, France
| | - Bernd Wissinger
- Institute for Ophthalmic Research, Centre for Ophthalmology, University of Tübingen, Tübingen, Germany
| | - Marion Szelechowski
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université Toulouse III, CNRS, Toulouse, France
| | - Pascale Belenguer
- Centre de Recherches sur la Cognition Animale (CRCA), Centre de Biologie Intégrative (CBI), Université Toulouse III, CNRS, Toulouse, France
| |
Collapse
|
5
|
Pohl KA, Zhang X, Pham AH, Chan JW, Sadun AA, Yang XJ. Establishing induced pluripotent stem cell lines from two dominant optic atrophy patients with distinct OPA1 mutations and clinical pathologies. Front Genet 2023; 14:1251216. [PMID: 37745862 PMCID: PMC10513078 DOI: 10.3389/fgene.2023.1251216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/17/2023] [Indexed: 09/26/2023] Open
Abstract
Dominant optic atrophy (DOA) is an inherited disease that leads to the loss of retinal ganglion cells (RGCs), the projection neurons that relay visual information from the retina to the brain through the optic nerve. The majority of DOA cases can be attributed to mutations in optic atrophy 1 (OPA1), a nuclear gene encoding a mitochondrial-targeted protein that plays important roles in maintaining mitochondrial structure, dynamics, and bioenergetics. Although OPA1 is ubiquitously expressed in all human tissues, RGCs appear to be the primary cell type affected by OPA1 mutations. DOA has not been extensively studied in human RGCs due to the general unavailability of retinal tissues. However, recent advances in stem cell biology have made it possible to produce human RGCs from pluripotent stem cells (PSCs). To aid in establishing DOA disease models based on human PSC-derived RGCs, we have generated iPSC lines from two DOA patients who carry distinct OPA1 mutations and present very different disease symptoms. Studies using these OPA1 mutant RGCs can be correlated with clinical features in the patients to provide insights into DOA disease mechanisms.
Collapse
Affiliation(s)
- Katherine A. Pohl
- Department of Ophthalmology, Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Xiangmei Zhang
- Department of Ophthalmology, Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA, United States
| | - Anh H. Pham
- Department of Ophthalmology, Doheny Eye Institute, University of California, Los Angeles, Pasadena, CA, United States
| | - Jane W. Chan
- Department of Ophthalmology, Doheny Eye Institute, University of California, Los Angeles, Pasadena, CA, United States
| | - Alfredo A. Sadun
- Department of Ophthalmology, Doheny Eye Institute, University of California, Los Angeles, Pasadena, CA, United States
| | - Xian-Jie Yang
- Department of Ophthalmology, Stein Eye Institute, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
6
|
Tribble JR, Hui F, Quintero H, El Hajji S, Bell K, Di Polo A, Williams PA. Neuroprotection in glaucoma: Mechanisms beyond intraocular pressure lowering. Mol Aspects Med 2023; 92:101193. [PMID: 37331129 DOI: 10.1016/j.mam.2023.101193] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/25/2023] [Accepted: 06/04/2023] [Indexed: 06/20/2023]
Abstract
Glaucoma is a common, complex, multifactorial neurodegenerative disease characterized by progressive dysfunction and then loss of retinal ganglion cells, the output neurons of the retina. Glaucoma is the most common cause of irreversible blindness and affects ∼80 million people worldwide with many more undiagnosed. The major risk factors for glaucoma are genetics, age, and elevated intraocular pressure. Current strategies only target intraocular pressure management and do not directly target the neurodegenerative processes occurring at the level of the retinal ganglion cell. Despite strategies to manage intraocular pressure, as many as 40% of glaucoma patients progress to blindness in at least one eye during their lifetime. As such, neuroprotective strategies that target the retinal ganglion cell and these neurodegenerative processes directly are of great therapeutic need. This review will cover the recent advances from basic biology to on-going clinical trials for neuroprotection in glaucoma covering degenerative mechanisms, metabolism, insulin signaling, mTOR, axon transport, apoptosis, autophagy, and neuroinflammation. With an increased understanding of both the basic and clinical mechanisms of the disease, we are closer than ever to a neuroprotective strategy for glaucoma.
Collapse
Affiliation(s)
- James R Tribble
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Flora Hui
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, Melbourne, Australia; Department of Optometry & Vision Sciences, The University of Melbourne, Melbourne, Australia
| | - Heberto Quintero
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Sana El Hajji
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Katharina Bell
- NHMRC Clinical Trials Centre, University of Sydney, Australia; Eye ACP Duke-NUS, Singapore
| | - Adriana Di Polo
- Department of Neuroscience, University of Montreal, Montreal, Canada; Neuroscience Division, Centre de recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Montreal, Canada
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
7
|
Wang L, Yang Z, He X, Pu S, Yang C, Wu Q, Zhou Z, Cen X, Zhao H. Mitochondrial protein dysfunction in pathogenesis of neurological diseases. Front Mol Neurosci 2022; 15:974480. [PMID: 36157077 PMCID: PMC9489860 DOI: 10.3389/fnmol.2022.974480] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 08/08/2022] [Indexed: 11/21/2022] Open
Abstract
Mitochondria are essential organelles for neuronal function and cell survival. Besides the well-known bioenergetics, additional mitochondrial roles in calcium signaling, lipid biogenesis, regulation of reactive oxygen species, and apoptosis are pivotal in diverse cellular processes. The mitochondrial proteome encompasses about 1,500 proteins encoded by both the nuclear DNA and the maternally inherited mitochondrial DNA. Mutations in the nuclear or mitochondrial genome, or combinations of both, can result in mitochondrial protein deficiencies and mitochondrial malfunction. Therefore, mitochondrial quality control by proteins involved in various surveillance mechanisms is critical for neuronal integrity and viability. Abnormal proteins involved in mitochondrial bioenergetics, dynamics, mitophagy, import machinery, ion channels, and mitochondrial DNA maintenance have been linked to the pathogenesis of a number of neurological diseases. The goal of this review is to give an overview of these pathways and to summarize the interconnections between mitochondrial protein dysfunction and neurological diseases.
Collapse
Affiliation(s)
- Liang Wang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Ziyun Yang
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital of Sichuan University, Chengdu, China
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Xiumei He
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Shiming Pu
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Cheng Yang
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Qiong Wu
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Zuping Zhou
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
| | - Xiaobo Cen
- National Chengdu Center for Safety Evaluation of Drugs, State Key Laboratory of Biotherapy/Collaborative Innovation Center for Biotherapy, West China Hospital of Sichuan University, Chengdu, China
| | - Hongxia Zhao
- School of Life Sciences, Guangxi Normal University, Guilin, China
- Guangxi Universities, Key Laboratory of Stem Cell and Biopharmaceutical Technology, Guangxi Normal University, Guilin, China
- Research Center for Biomedical Sciences, Guangxi Normal University, Guilin, China
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| |
Collapse
|
8
|
Tian R, Guo H, Jin Z, Zhang F, Zhao J, Seim I. Molecular evolution of vision-related genes may contribute to marsupial photic niche adaptations. Front Ecol Evol 2022. [DOI: 10.3389/fevo.2022.982073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Vision plays an essential role in the life of many animals. While most mammals are night-active (nocturnal), many have adapted to novel light environments. This includes diurnal (day-active) and crepuscular (twilight-active) species. Here, we used integrative approaches to investigate the molecular evolution of 112 vision-related genes across 19 genomes representing most marsupial orders. We found that four genes (GUCA1B, GUCY2F, RGR, and SWS2) involved in retinal phototransduction likely became functionally redundant in the ancestor of marsupials, a group of largely obligate nocturnal mammals. We also show evidence of rapid evolution and positive selection of bright-light vision genes in the common ancestor of Macropus (kangaroos, wallaroos, and wallabies). Macropus-specific amino acid substitutions in opsin genes (LWS and SWS1), in particular, may be an adaptation for crepuscular vision in this genus via opsin spectral sensitivity tuning. Our study set the stage for functional genetics studies and provides a stepping stone to future research efforts that fully capture the visual repertoire of marsupials.
Collapse
|
9
|
Zaninello M, Palikaras K, Sotiriou A, Tavernarakis N, Scorrano L. Sustained intracellular calcium rise mediates neuronal mitophagy in models of autosomal dominant optic atrophy. Cell Death Differ 2022; 29:167-177. [PMID: 34389813 PMCID: PMC8738763 DOI: 10.1038/s41418-021-00847-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial dysfunction and mitophagy are often hallmarks of neurodegenerative diseases such as autosomal dominant optic atrophy (ADOA) caused by mutations in the key mitochondrial dynamics protein optic atrophy 1 (Opa1). However, the second messengers linking mitochondrial dysfunction to initiation of mitophagy remain poorly characterized. Here, we show in mammalian and nematode neurons that Opa1 mutations trigger Ca2+-dependent mitophagy. Deletion or expression of mutated Opa1 in mouse retinal ganglion cells and Caenorhabditis elegans motor neurons lead to mitochondrial dysfunction, increased cytosolic Ca2+ levels, and decreased axonal mitochondrial density. Chelation of Ca2+ restores mitochondrial density in neuronal processes, neuronal function, and viability. Mechanistically, sustained Ca2+ levels activate calcineurin and AMPK, placed in the same genetic pathway regulating axonal mitochondrial density. Our data reveal that mitophagy in ADOA depends on Ca2+-calcineurin-AMPK signaling cascade.
Collapse
Affiliation(s)
- Marta Zaninello
- grid.428736.cVeneto Institute of Molecular Medicine, Padova, Italy ,grid.5608.b0000 0004 1757 3470Department of Biology, University of Padova, Padova, Italy ,grid.6190.e0000 0000 8580 3777Present Address: Cologne Excellence Cluster on Cellular Stress Responses in Aging‐Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Konstantinos Palikaras
- grid.5216.00000 0001 2155 0800Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Aggeliki Sotiriou
- grid.4834.b0000 0004 0635 685XInstitute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete Greece
| | - Nektarios Tavernarakis
- grid.4834.b0000 0004 0635 685XInstitute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Crete Greece ,grid.8127.c0000 0004 0576 3437Department of Basic Sciences, School of Medicine, University of Crete, Heraklion, Crete Greece
| | - Luca Scorrano
- grid.428736.cVeneto Institute of Molecular Medicine, Padova, Italy ,grid.5608.b0000 0004 1757 3470Department of Biology, University of Padova, Padova, Italy
| |
Collapse
|
10
|
Tang J, Zhuo Y, Li Y. Effects of Iron and Zinc on Mitochondria: Potential Mechanisms of Glaucomatous Injury. Front Cell Dev Biol 2021; 9:720288. [PMID: 34447755 PMCID: PMC8383321 DOI: 10.3389/fcell.2021.720288] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 07/22/2021] [Indexed: 12/26/2022] Open
Abstract
Glaucoma is the most substantial cause of irreversible blinding, which is accompanied by progressive retinal ganglion cell damage. Retinal ganglion cells are energy-intensive neurons that connect the brain and retina, and depend on mitochondrial homeostasis to transduce visual information through the brain. As cofactors that regulate many metabolic signals, iron and zinc have attracted increasing attention in studies on neurons and neurodegenerative diseases. Here, we summarize the research connecting iron, zinc, neuronal mitochondria, and glaucomatous injury, with the aim of updating and expanding the current view of how retinal ganglion cells degenerate in glaucoma, which can reveal novel potential targets for neuroprotection.
Collapse
Affiliation(s)
- Jiahui Tang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yehong Zhuo
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yiqing Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
11
|
Muench NA, Patel S, Maes ME, Donahue RJ, Ikeda A, Nickells RW. The Influence of Mitochondrial Dynamics and Function on Retinal Ganglion Cell Susceptibility in Optic Nerve Disease. Cells 2021; 10:cells10071593. [PMID: 34201955 PMCID: PMC8306483 DOI: 10.3390/cells10071593] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Revised: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/30/2022] Open
Abstract
The important roles of mitochondrial function and dysfunction in the process of neurodegeneration are widely acknowledged. Retinal ganglion cells (RGCs) appear to be a highly vulnerable neuronal cell type in the central nervous system with respect to mitochondrial dysfunction but the actual reasons for this are still incompletely understood. These cells have a unique circumstance where unmyelinated axons must bend nearly 90° to exit the eye and then cross a translaminar pressure gradient before becoming myelinated in the optic nerve. This region, the optic nerve head, contains some of the highest density of mitochondria present in these cells. Glaucoma represents a perfect storm of events occurring at this location, with a combination of changes in the translaminar pressure gradient and reassignment of the metabolic support functions of supporting glia, which appears to apply increased metabolic stress to the RGC axons leading to a failure of axonal transport mechanisms. However, RGCs themselves are also extremely sensitive to genetic mutations, particularly in genes affecting mitochondrial dynamics and mitochondrial clearance. These mutations, which systemically affect the mitochondria in every cell, often lead to an optic neuropathy as the sole pathologic defect in affected patients. This review summarizes knowledge of mitochondrial structure and function, the known energy demands of neurons in general, and places these in the context of normal and pathological characteristics of mitochondria attributed to RGCs.
Collapse
Affiliation(s)
- Nicole A. Muench
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
| | - Sonia Patel
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
| | - Margaret E. Maes
- Institute of Science and Technology Austria, 3400 Klosterneuburg, Austria;
| | - Ryan J. Donahue
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
- Boston Children’s Hospital, Harvard Medical School, Harvard University, Boston, MA 02115, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA;
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Robert W. Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI 53706, USA; (N.A.M.); (S.P.); (R.J.D.)
- McPherson Eye Research Institute, University of Wisconsin-Madison, Madison, WI 53705, USA
- Correspondence:
| |
Collapse
|
12
|
Del Dotto V, Carelli V. Dominant Optic Atrophy (DOA): Modeling the Kaleidoscopic Roles of OPA1 in Mitochondrial Homeostasis. Front Neurol 2021; 12:681326. [PMID: 34177786 PMCID: PMC8220150 DOI: 10.3389/fneur.2021.681326] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/22/2021] [Indexed: 02/05/2023] Open
Abstract
In the year 2000, the discovery of OPA1 mutations as causative for dominant optic atrophy (DOA) was pivotal to rapidly expand the field of mitochondrial dynamics and describe the complex machinery governing this pathway, with a multitude of other genes and encoded proteins involved in neurodegenerative disorders of the optic nerve. OPA1 turned out to be a much more complex protein than initially envisaged, connecting multiple pathways beyond its strict role in mitochondrial fusion, such as sensing of OXPHOS needs and mitochondrial DNA maintenance. As a consequence, an increasing need to investigate OPA1 functions at multiple levels has imposed the development of multiple tools and models that are here reviewed. Translational mitochondrial medicine, with the ultimate objective of translating basic science necessary to understand pathogenic mechanisms into therapeutic strategies, requires disease modeling at multiple levels: from the simplest, like in yeast, to cell models, including the increasing use of reprogrammed stem cells (iPSCs) from patients, to animal models. In the present review, we thoroughly examine and provide the state of the art of all these approaches.
Collapse
Affiliation(s)
- Valentina Del Dotto
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Valerio Carelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy.,Istituto di Ricovero e Cura a Carattere Scientifico Istituto delle Scienze Neurologiche di Bologna, Programma di Neurogenetica, Bologna, Italy
| |
Collapse
|
13
|
Liu H, Prokosch V. Energy Metabolism in the Inner Retina in Health and Glaucoma. Int J Mol Sci 2021; 22:ijms22073689. [PMID: 33916246 PMCID: PMC8036449 DOI: 10.3390/ijms22073689] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 02/07/2023] Open
Abstract
Glaucoma, the leading cause of irreversible blindness, is a heterogeneous group of diseases characterized by progressive loss of retinal ganglion cells (RGCs) and their axons and leads to visual loss and blindness. Risk factors for the onset and progression of glaucoma include systemic and ocular factors such as older age, lower ocular perfusion pressure, and intraocular pressure (IOP). Early signs of RGC damage comprise impairment of axonal transport, downregulation of specific genes and metabolic changes. The brain is often cited to be the highest energy-demanding tissue of the human body. The retina is estimated to have equally high demands. RGCs are particularly active in metabolism and vulnerable to energy insufficiency. Understanding the energy metabolism of the inner retina, especially of the RGCs, is pivotal for understanding glaucoma’s pathophysiology. Here we review the key contributors to the high energy demands in the retina and the distinguishing features of energy metabolism of the inner retina. The major features of glaucoma include progressive cell death of retinal ganglions and optic nerve damage. Therefore, this review focuses on the energetic budget of the retinal ganglion cells, optic nerve and the relevant cells that surround them.
Collapse
|
14
|
Li M, Guo J, Wang H, Li Y. Involvement of Mitochondrial Dynamics and Mitophagy in Sevoflurane-Induced Cell Toxicity. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6685468. [PMID: 33728028 PMCID: PMC7937461 DOI: 10.1155/2021/6685468] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 02/07/2023]
Abstract
General anesthesia is a powerful and indispensable tool to ensure the accomplishment of surgical procedures or clinical examinations. Sevoflurane as an inhalational anesthetic without unpleasant odor is commonly used in clinical practice, especially for pediatric surgery. However, the toxicity caused by sevoflurane has gained growing attention. Mitochondria play a key role in maintaining cellular metabolism and survival. To maintain the stability of mitochondrial homeostasis, they are constantly going through fusion and fission. Also, damaged mitochondria need to be degraded by autophagy, termed as mitophagy. Accumulating evidence proves that sevoflurane exposure in young age could lead to cell toxicity by triggering the mitochondrial pathway of apoptosis, inducing the abnormalities of mitochondrial dynamics and mitophagy. In the present review, we focus on the current understanding of mitochondrial apoptosis, dynamics and mitophagy in cell function, the implications for cell toxicity in response to sevoflurane, and their underlying potential mechanisms.
Collapse
Affiliation(s)
- Ming Li
- School of Basic Medical Sciences, Hebei University, Baoding, Hebei Province, China
| | - Jiguang Guo
- School of Basic Medical Sciences, Hebei University, Baoding, Hebei Province, China
| | - Hongjie Wang
- School of Basic Medical Sciences, Hebei University, Baoding, Hebei Province, China
- Affiliated Hospital of Hebei University, Baoding, Hebei Province, China
| | - Yuzhen Li
- Department of Pathophysiology, Graduate School of PLA General Hospital, Beijing, China
| |
Collapse
|
15
|
Ng WSV, Trigano M, Freeman T, Varrichio C, Kandaswamy DK, Newland B, Brancale A, Rozanowska M, Votruba M. New avenues for therapy in mitochondrial optic neuropathies. THERAPEUTIC ADVANCES IN RARE DISEASE 2021; 2:26330040211029037. [PMID: 37181108 PMCID: PMC10032437 DOI: 10.1177/26330040211029037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 06/10/2021] [Indexed: 05/16/2023]
Abstract
Mitochondrial optic neuropathies are a group of optic nerve atrophies exemplified by the two commonest conditions in this group, autosomal dominant optic atrophy (ADOA) and Leber's hereditary optic neuropathy (LHON). Their clinical features comprise reduced visual acuity, colour vision deficits, centro-caecal scotomas and optic disc pallor with thinning of the retinal nerve fibre layer. The primary aetiology is genetic, with underlying nuclear or mitochondrial gene mutations. The primary pathology is owing to retinal ganglion cell dysfunction and degeneration. There is currently only one approved treatment and no curative therapy is available. In this review we summarise the genetic and clinical features of ADOA and LHON and then examine what new avenues there may be for therapeutic intervention. The therapeutic strategies to manage LHON and ADOA can be split into four categories: prevention, compensation, replacement and repair. Prevention is technically an option by modifying risk factors such as smoking cessation, or by utilising pre-implantation genetic diagnosis, although this is unlikely to be applied in mitochondrial optic neuropathies due to the non-life threatening and variable nature of these conditions. Compensation involves pharmacological interventions that ameliorate the mitochondrial dysfunction at a cellular and tissue level. Replacement and repair are exciting new emerging areas. Clinical trials, both published and underway, in this area are likely to reveal future potential benefits, since new therapies are desperately needed. Plain language summary Optic nerve damage leading to loss of vision can be caused by a variety of insults. One group of conditions leading to optic nerve damage is caused by defects in genes that are essential for cells to make energy in small organelles called mitochondria. These conditions are known as mitochondrial optic neuropathies and two predominant examples are called autosomal dominant optic atrophy and Leber's hereditary optic neuropathy. Both conditions are caused by problems with the energy powerhouse of cells: mitochondria. The cells that are most vulnerable to this mitochondrial malfunction are called retinal ganglion cells, otherwise collectively known as the optic nerve, and they take the electrical impulse from the retina in the eye to the brain. The malfunction leads to death of some of the optic nerve cells, the degree of vision loss being linked to the number of those cells which are impacted in this way. Patients will lose visual acuity and colour vision and develop a central blind spot in their field of vision. There is currently no cure and very few treatment options. New treatments are desperately needed for patients affected by these devastating diseases. New treatments can potentially arise in four ways: prevention, compensation, replacement and repair of the defects. Here we explore how present and possible future treatments might provide hope for those suffering from these conditions.
Collapse
Affiliation(s)
| | - Matthieu Trigano
- Mitochondria and Vision Lab, School of
Optometry and Vision Sciences, Cardiff University, Cardiff, UK
| | - Thomas Freeman
- Mitochondria and Vision Lab, School of
Optometry and Vision Sciences, Cardiff University, Cardiff, UK
| | - Carmine Varrichio
- School of Pharmacy and Pharmaceutical Sciences,
Cardiff University, Cardiff, UK
| | - Dinesh Kumar Kandaswamy
- Mitochondria and Vision Lab, School of
Optometry and Vision Sciences, Cardiff University, Cardiff, UK
| | - Ben Newland
- School of Pharmacy and Pharmaceutical Sciences,
Cardiff University, Cardiff, UK
| | - Andrea Brancale
- School of Pharmacy and Pharmaceutical Sciences,
Cardiff University, Cardiff, UK
| | - Malgorzata Rozanowska
- Mitochondria and Vision Lab, School of
Optometry and Vision Sciences, Cardiff University, Cardiff, UK
| | - Marcela Votruba
- School of Optometry and Vision Sciences,
Cardiff University, Maindy Road, Cardiff, CF24 4HQ, Wales, UK; Cardiff Eye
Unit, University Hospital of Wales, Cardiff, UK
| |
Collapse
|
16
|
Cimaglia G, Votruba M, Morgan JE, André H, Williams PA. Potential Therapeutic Benefit of NAD + Supplementation for Glaucoma and Age-Related Macular Degeneration. Nutrients 2020; 12:nu12092871. [PMID: 32961812 PMCID: PMC7551676 DOI: 10.3390/nu12092871] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 09/14/2020] [Accepted: 09/17/2020] [Indexed: 12/13/2022] Open
Abstract
Glaucoma and age-related macular degeneration are leading causes of irreversible blindness worldwide with significant health and societal burdens. To date, no clinical cures are available and treatments target only the manageable symptoms and risk factors (but do not remediate the underlying pathology of the disease). Both diseases are neurodegenerative in their pathology of the retina and as such many of the events that trigger cell dysfunction, degeneration, and eventual loss are due to mitochondrial dysfunction, inflammation, and oxidative stress. Here, we critically review how a decreased bioavailability of nicotinamide adenine dinucleotide (NAD; a crucial metabolite in healthy and disease states) may underpin many of these aberrant mechanisms. We propose how exogenous sources of NAD may become a therapeutic standard for the treatment of these conditions.
Collapse
Affiliation(s)
- Gloria Cimaglia
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 112 82 Stockholm, Sweden;
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, Wales, UK; (M.V.); (J.E.M.)
| | - Marcela Votruba
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, Wales, UK; (M.V.); (J.E.M.)
- Cardiff Eye Unit, University Hospital Wales, Cardiff CF14 4XW, Wales, UK
| | - James E. Morgan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4HQ, Wales, UK; (M.V.); (J.E.M.)
- School of Medicine, Cardiff University, Cardiff CF14 4YS, Wales, UK
| | - Helder André
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 112 82 Stockholm, Sweden;
- Correspondence: (H.A.); (P.A.W.)
| | - Pete A. Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 112 82 Stockholm, Sweden;
- Correspondence: (H.A.); (P.A.W.)
| |
Collapse
|
17
|
Retinal energy metabolism in health and glaucoma. Prog Retin Eye Res 2020; 81:100881. [PMID: 32712136 DOI: 10.1016/j.preteyeres.2020.100881] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/25/2020] [Accepted: 06/28/2020] [Indexed: 01/17/2023]
Abstract
Energy metabolism refers to the processes by which life transfers energy to do cellular work. The retina's relatively large energy demands make it vulnerable to energy insufficiency. In addition, evolutionary pressures to optimize human vision have been traded against retinal ganglion cell bioenergetic fragility. Details of the metabolic profiles of the different retinal cells remain poorly understood and are challenging to resolve. Detailed immunohistochemical mapping of the energy pathway enzymes and substrate transporters has provided some insights and highlighted interspecies differences. The different spatial metabolic patterns between the vascular and avascular retinas can account for some inconsistent data in the literature. There is a consilience of evidence that at least some individuals with glaucoma have impaired RGC energy metabolism, either due to impaired nutrient supply or intrinsic metabolic perturbations. Bioenergetic-based therapy for glaucoma has a compelling pathophysiological foundation and is supported by recent successes in animal models. Recent demonstrations of visual and electrophysiological neurorecovery in humans with glaucoma is highly encouraging and motivates longer duration trials investigating bioenergetic neuroprotection.
Collapse
|
18
|
Bevan RJ, Williams PA, Waters CT, Thirgood R, Mui A, Seto S, Good M, Morgan JE, Votruba M, Erchova I. OPA1 deficiency accelerates hippocampal synaptic remodelling and age-related deficits in learning and memory. Brain Commun 2020; 2:fcaa101. [PMID: 33094281 PMCID: PMC7566495 DOI: 10.1093/braincomms/fcaa101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 04/09/2020] [Accepted: 05/27/2020] [Indexed: 12/13/2022] Open
Abstract
A healthy mitochondrial network is essential for the maintenance of neuronal synaptic integrity. Mitochondrial and metabolic dysfunction contributes to the pathogenesis of many neurodegenerative diseases including dementia. OPA1 is the master regulator of mitochondrial fusion and fission and is likely to play an important role during neurodegenerative events. To explore this, we quantified hippocampal dendritic and synaptic integrity and the learning and memory performance of aged Opa1 haploinsufficient mice carrying the Opa1Q285X mutation (B6; C3-Opa1Q285STOP ; Opa1+/- ). We demonstrate that heterozygous loss of Opa1 results in premature age-related loss of spines in hippocampal pyramidal CA1 neurons and a reduction in synaptic density in the hippocampus. This loss is associated with subtle memory deficits in both spatial novelty and object recognition. We hypothesize that metabolic failure to maintain normal neuronal activity at the level of a single spine leads to premature age-related memory deficits. These results highlight the importance of mitochondrial homeostasis for maintenance of neuronal function during ageing.
Collapse
Affiliation(s)
- Ryan J Bevan
- School of Optometry and Vision Sciences, Cardiff University, Maindy Rd, Cardiff, CF24 4HQ, UK
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, Polhemsgatan 50, 112 82 Stockholm, Sweden
| | - Caroline T Waters
- School of Optometry and Vision Sciences, Cardiff University, Maindy Rd, Cardiff, CF24 4HQ, UK
| | - Rebecca Thirgood
- School of Optometry and Vision Sciences, Cardiff University, Maindy Rd, Cardiff, CF24 4HQ, UK
| | - Amanda Mui
- School of Optometry and Vision Sciences, Cardiff University, Maindy Rd, Cardiff, CF24 4HQ, UK
| | - Sharon Seto
- School of Optometry and Vision Sciences, Cardiff University, Maindy Rd, Cardiff, CF24 4HQ, UK
| | - Mark Good
- School of Psychology, Cardiff University, Tower Building, 70 Park Place, Cardiff, CF10 3AT, UK
| | - James E Morgan
- School of Optometry and Vision Sciences, Cardiff University, Maindy Rd, Cardiff, CF24 4HQ, UK
| | - Marcela Votruba
- School of Optometry and Vision Sciences, Cardiff University, Maindy Rd, Cardiff, CF24 4HQ, UK
| | - Irina Erchova
- School of Optometry and Vision Sciences, Cardiff University, Maindy Rd, Cardiff, CF24 4HQ, UK
| |
Collapse
|
19
|
Abstract
Mitochondrial dysfunction or loss is evident in neurodegenerative diseases. Furthermore, mitochondrial DNA (mtDNA) mutations associated with NADH dehydrogenase subunits and nuclear gene mutations that affect mitochondrial function result in optic neuropathies. In this issue of the JCI, Del Dotto et al. and Piro-Mégy et al. identify heterozygous mutations in nuclear-encoded mitochondrial single-strand binding protein 1 (SSBP1) in patients with apparently dominant optic neuropathy with or without extraocular phenotypes. Both research groups reported similar mitochondrial findings in response to SSBP1 mutations. However, the specific SSBP1 mitochondria-associated function in retinal ganglion cells (RGCs) and the resulting optic nerve remains unclear. We suggest that high expression of SSBP1 during RGC differentiation is critical for mtDNA maintenance to produce appropriate optic nerve connectivity and that SSBP1 mutations in dominant optic atrophy patients do not permit stable binding to N6-methyldeoxyadenosine on the heavy strand involved with replication, leading to disruptions of mtDNA and, eventually, optic nerve dysfunction.
Collapse
|
20
|
Tribble JR, Vasalauskaite A, Redmond T, Young RD, Hassan S, Fautsch MP, Sengpiel F, Williams PA, Morgan JE. Midget retinal ganglion cell dendritic and mitochondrial degeneration is an early feature of human glaucoma. Brain Commun 2019; 1:fcz035. [PMID: 31894207 PMCID: PMC6928391 DOI: 10.1093/braincomms/fcz035] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 10/29/2019] [Accepted: 11/01/2019] [Indexed: 12/31/2022] Open
Abstract
Glaucoma is characterized by the progressive dysfunction and loss of retinal ganglion cells. However, the earliest degenerative events that occur in human glaucoma are relatively unknown. Work in animal models has demonstrated that retinal ganglion cell dendrites remodel and atrophy prior to the loss of the cell soma. Whether this occurs in human glaucoma has yet to be elucidated. Serial block face scanning electron microscopy is well established as a method to determine neuronal connectivity at high resolution but so far has only been performed in normal retina from animal models. To assess the structure-function relationship of early human glaucomatous neurodegeneration, regions of inner retina assessed to have none-to-moderate loss of retinal ganglion cell number were processed using serial block face scanning electron microscopy (n = 4 normal retinas, n = 4 glaucoma retinas). This allowed detailed 3D reconstruction of retinal ganglion cells and their intracellular components at a nanometre scale. In our datasets, retinal ganglion cell dendrites degenerate early in human glaucoma, with remodelling and redistribution of the mitochondria. We assessed the relationship between visual sensitivity and retinal ganglion cell density and discovered that this only partially conformed to predicted models of structure-function relationships, which may be affected by these early neurodegenerative changes. In this study, human glaucomatous retinal ganglion cells demonstrate compartmentalized degenerative changes as observed in animal models. Importantly, in these models, many of these changes have been demonstrated to be reversible, increasing the likelihood of translation to viable therapies for human glaucoma.
Collapse
Affiliation(s)
- James R Tribble
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, CF24 4HQ Wales, UK
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 112 82 Stockholm, Sweden
| | | | - Tony Redmond
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, CF24 4HQ Wales, UK
| | - Robert D Young
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, CF24 4HQ Wales, UK
| | - Shoaib Hassan
- School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XW Wales, UK
| | | | - Frank Sengpiel
- School of Biosciences, Cardiff University, Cardiff, CF10 3AX Wales, UK
| | - Pete A Williams
- Department of Clinical Neuroscience, Division of Eye and Vision, St. Erik Eye Hospital, Karolinska Institutet, 112 82 Stockholm, Sweden
| | - James E Morgan
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, CF24 4HQ Wales, UK
- School of Medicine, Cardiff University, Heath Park, Cardiff, CF14 4XW Wales, UK
| |
Collapse
|
21
|
Tao JX, Zhou WC, Zhu XG. Mitochondria as Potential Targets and Initiators of the Blue Light Hazard to the Retina. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:6435364. [PMID: 31531186 PMCID: PMC6721470 DOI: 10.1155/2019/6435364] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 06/18/2019] [Accepted: 07/25/2019] [Indexed: 12/20/2022]
Abstract
Commercially available white light-emitting diodes (LEDs) have an intense emission in the range of blue light, which has raised a range of public concerns about their potential risks as retinal hazards. Distinct from other visible light components, blue light is characterized by short wavelength, high energy, and strong penetration that can reach the retina with relatively little loss in damage potential. Mitochondria are abundant in retinal tissues, giving them relatively high access to blue light, and chromophores, which are enriched in the retina, have many mitochondria able to absorb blue light and induce photochemical effects. Therefore, excessive exposure of the retina to blue light tends to cause ROS accumulation and oxidative stress, which affect the structure and function of the retinal mitochondria and trigger mitochondria-involved death signaling pathways. In this review, we highlight the essential roles of mitochondria in blue light-induced photochemical damage and programmed cell death in the retina, indicate directions for future research and preventive targets in terms of the blue light hazard to the retina, and suggest applying LED devices in a rational way to prevent the blue light hazard.
Collapse
Affiliation(s)
- Jin-Xin Tao
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Department of Clinical Medicine, The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Wen-Chuan Zhou
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
- Department of Clinical Medicine, The Second Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Xin-Gen Zhu
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang 330006, China
| |
Collapse
|
22
|
The Metabolomic Signature of Opa1 Deficiency in Rat Primary Cortical Neurons Shows Aspartate/Glutamate Depletion and Phospholipids Remodeling. Sci Rep 2019; 9:6107. [PMID: 30988455 PMCID: PMC6465244 DOI: 10.1038/s41598-019-42554-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Accepted: 03/26/2019] [Indexed: 12/25/2022] Open
Abstract
Pathogenic variants of OPA1, which encodes a dynamin GTPase involved in mitochondrial fusion, are responsible for a spectrum of neurological disorders sharing optic nerve atrophy and visual impairment. To gain insight on OPA1 neuronal specificity, we performed targeted metabolomics on rat cortical neurons with OPA1 expression inhibited by RNA interference. Of the 103 metabolites accurately measured, univariate analysis including the Benjamini-Hochberg correction revealed 6 significantly different metabolites in OPA1 down-regulated neurons, with aspartate being the most significant (p < 0.001). Supervised multivariate analysis by OPLS-DA yielded a model with good predictive capability (Q2cum = 0.65) and a low risk of over-fitting (permQ2 = -0.16, CV-ANOVA p-value 0.036). Amongst the 46 metabolites contributing the most to the metabolic signature were aspartate, glutamate and threonine, which all decreased in OPA1 down-regulated neurons, and lysine, 4 sphingomyelins, 4 lysophosphatidylcholines and 32 phosphatidylcholines which were increased. The phospholipid signature may reflect intracellular membrane remodeling due to loss of mitochondrial fusion and/or lipid droplet accumulation. Aspartate and glutamate deficiency, also found in the plasma of OPA1 patients, is likely the consequence of respiratory chain deficiency, whereas the glutamate decrease could contribute to the synaptic dysfunction that we previously identified in this model.
Collapse
|
23
|
Mitochondrial pathophysiology beyond the retinal ganglion cell: occipital GABA is decreased in autosomal dominant optic neuropathy. Graefes Arch Clin Exp Ophthalmol 2018; 256:2341-2348. [PMID: 30324419 PMCID: PMC6224020 DOI: 10.1007/s00417-018-4153-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 07/05/2018] [Accepted: 09/24/2018] [Indexed: 01/05/2023] Open
Abstract
PURPOSE It has remained a mystery why some genetic mitochondrial disorders affect predominantly specific cell types such as the retinal ganglion cell. This is particularly intriguing concerning retinal and cortical function since they are tightly linked in health and disease. Autosomal dominant optic neuropathy (ADOA) is a mitochondrial disease that affects the ganglion cell. However, it is unknown whether alterations are also present in the visual cortex, namely in excitation/inhibition balance. METHODS In this study, we performed in vivo structural and biochemical proton magnetic resonance imaging in 14 ADOA and 11 age-matched control participants focusing on the visual cortex, with the aim of establishing whether in this genetically determined disease an independent cortical neurochemical phenotype could be established irrespective of a putative structural phenotype. Cortical thickness of anatomically defined visual areas was estimated, and a voxel-based morphometry approach was used to assess occipital volumetric changes in ADOA. Neurochemical measurements were focused on γ-aminobutyric acid (GABA) and glutamate, as indicators of the local excitatory/inhibitory balance. RESULTS We found evidence for reduced visual cortical GABA and preserved glutamate concentrations in the absence of cortical or subcortical atrophy. These changes in GABA levels were explained by neither structural nor functional measures of visual loss, suggesting a developmental origin. CONCLUSIONS These results suggest that mitochondrial disorders that were previously believed to only affect retinal function may also affect cortical physiology, especially the GABAergic system, suggesting reduced brain inhibition vs. excitation. This GABA phenotype, independent of sensory loss or cortical atrophy and in the presence of preserved glutamate levels, suggests a neurochemical developmental change at the cortical level, leading to a pathophysiological excitation/inhibition imbalance.
Collapse
|
24
|
Diot A, Agnew T, Sanderson J, Liao C, Carver J, Neves RPD, Gupta R, Guo Y, Waters C, Seto S, Daniels MJ, Dombi E, Lodge T, Morten K, Williams SA, Enver T, Iborra FJ, Votruba M, Poulton J. Validating the RedMIT/GFP-LC3 Mouse Model by Studying Mitophagy in Autosomal Dominant Optic Atrophy Due to the OPA1Q285STOP Mutation. Front Cell Dev Biol 2018; 6:103. [PMID: 30283778 PMCID: PMC6156146 DOI: 10.3389/fcell.2018.00103] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 08/13/2018] [Indexed: 01/02/2023] Open
Abstract
Background: Autosomal dominant optic atrophy (ADOA) is usually caused by mutations in the essential gene, OPA1. This encodes a ubiquitous protein involved in mitochondrial dynamics, hence tissue specificity is not understood. Dysregulated mitophagy (mitochondria recycling) is implicated in ADOA, being increased in OPA1 patient fibroblasts. Furthermore, autophagy may be increased in retinal ganglion cells (RGCs) of the OPA1Q285STOP mouse model. Aims: We developed a mouse model for studying mitochondrial dynamics in order to investigate mitophagy in ADOA. Methods: We crossed the OPA1Q285STOP mouse with our RedMIT/GFP-LC3 mouse, harboring red fluorescent mitochondria and green fluorescent autophagosomes. Colocalization between mitochondria and autophagosomes, the hallmark of mitophagy, was quantified in fluorescently labeled organelles in primary cell cultures, using two high throughput imaging methods Imagestream (Amnis) and IN Cell Analyzer 1000 (GE Healthcare Life Sciences). We studied colocalization between mitochondria and autophagosomes in fixed sections using confocal microscopy. Results: We validated our imaging methods for RedMIT/GFP-LC3 mouse cells, showing that colocalization of red fluorescent mitochondria and green fluorescent autophagosomes is a useful indicator of mitophagy. We showed that colocalization increases when lysosomal processing is impaired. Further, colocalization of mitochondrial fragments and autophagosomes is increased in cultures from the OPA1Q285STOP/RedMIT/GFP-LC3 mice compared to RedMIT/GFP-LC3 control mouse cells that were wild type for OPA1. This was apparent in both mouse embryonic fibroblasts (MEFs) using IN Cell 1000 and in splenocytes using ImageStream imaging flow cytometer (Amnis). We confirmed that this represents increased mitophagic flux using lysosomal inhibitors. We also used microscopy to investigate the level of mitophagy in the retina from the OPA1Q285STOP/RedMIT/GFP-LC3 mice and the RedMIT/GFP-LC3 control mice. However, the expression levels of fluorescent proteins and the image signal-to-background ratios precluded the detection of colocalization so we were unable to show any difference in colocalization between these mice. Conclusions: We show that colocalization of fluorescent mitochondria and autophagosomes in cell cultures, but not fixed tissues from the RedMIT/GFP-LC3, can be used to detect mitophagy. We used this model to confirm that mitophagy is increased in a mouse model of ADOA. It will be useful for cell based studies of diseases caused by impaired mitochondrial dynamics.
Collapse
Affiliation(s)
- Alan Diot
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Thomas Agnew
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Jeremy Sanderson
- Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Chunyan Liao
- Molecular Biology and Biotechnology, University of Sheffield, Sheffield, United Kingdom
| | - Janet Carver
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | | | - Rajeev Gupta
- UCL Cancer Institute, University College London, London, United Kingdom
| | - Yanping Guo
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Caroline Waters
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Sharon Seto
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Matthew J. Daniels
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, University of Oxford, Headington, United Kingdom
| | - Eszter Dombi
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Tiffany Lodge
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Karl Morten
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Suzannah A. Williams
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| | - Tariq Enver
- UCL Cancer Institute, University College London, London, United Kingdom
| | | | - Marcela Votruba
- School of Optometry and Vision Sciences, Cardiff University, Cardiff, United Kingdom
| | - Joanna Poulton
- Nuffield Department of Women's and Reproductive Health, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
25
|
Sheng W, Lu Y, Mei F, Wang N, Liu ZZ, Han YY, Wang HT, Zou S, Xu H, Zhang X. Effect of Resveratrol on Sirtuins, OPA1, and Fis1 Expression in Adult Zebrafish Retina. ACTA ACUST UNITED AC 2018; 59:4542-4551. [PMID: 30208422 DOI: 10.1167/iovs.18-24539] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Affiliation(s)
- Weiwei Sheng
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
- Queen Mary School of Nanchang University, Nanchang, China
| | - Ye Lu
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
| | - Feng Mei
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
| | - Ning Wang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
| | - Zhi-Zhi Liu
- Institute of Life Science of Nanchang University, Nanchang, China
- School of Life Sciences of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Ying-Ying Han
- Institute of Life Science of Nanchang University, Nanchang, China
- School of Life Sciences of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Han-Tsing Wang
- Institute of Life Science of Nanchang University, Nanchang, China
- School of Life Sciences of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Suqi Zou
- Institute of Life Science of Nanchang University, Nanchang, China
- School of Life Sciences of Nanchang University, Nanchang, China
| | - Hong Xu
- Institute of Life Science of Nanchang University, Nanchang, China
- School of Life Sciences of Nanchang University, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| | - Xu Zhang
- Affiliated Eye Hospital of Nanchang University, Jiangxi Research Institute of Ophthalmology and Visual Science, Nanchang, China
- Jiangxi Provincial Collaborative Innovation Center for Cardiovascular, Digestive and Neuropsychiatric Diseases, Nanchang, China
| |
Collapse
|
26
|
Zaninello M, Scorrano L. Rapidly purified ganglion cells from neonatal mouse retinas allow studies of mitochondrial morphology and autophagy. Pharmacol Res 2018; 138:16-24. [PMID: 30077733 DOI: 10.1016/j.phrs.2018.07.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Revised: 07/27/2018] [Accepted: 07/29/2018] [Indexed: 01/09/2023]
Abstract
Retinal explants and mixed primary cultures are currently used to investigate retinal ganglion cells (RGCs) pathophysiology and pharmacology, but information on yield, quality and quantity of contaminant cells for the available RGCs enrichment techniques is lacking. Here we compare two methods of mouse primary RGCs purification and show that mitochondrial and autophagy parameters can be measured in rapidly purified RGCs. RGCs were purified from P0 mouse eyes using two methods based on the surface antigen Thy1. In a two-step immunopanning purification, a subtraction plate bound macrophage antiserum removed contaminant macrophages and endothelial cells; unbound RGCs were then affinity selected using a plate-bound antiThy1 antibody. In an immunopanning-magnetic separation, macrophage-antiserum bound cells were first subtracted and then RGCs were positively selected using an antiThy1 antibody bound to a magnetic column. The two-steps immunopanning yielded low amounts of 90% pure RGCs, whereas RGCs represented 30% of the 6-fold more cells collected by immunopanning-magnetic separation. RGCs purified with both methods could be microelectroporated to image expressed mitochondria and autophagosomes fluorescent probes and to show that expression of pathogenic Optic atrophy 1 mutants causes mitochondrial fragmentation. Thus, these two methods purify primary mouse RGCs amenable to studies of cell morphology, mitochondrial biology and autophagy.
Collapse
Affiliation(s)
- Marta Zaninello
- Venetian Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy; Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy
| | - Luca Scorrano
- Venetian Institute of Molecular Medicine, Via Orus 2, 35129 Padova, Italy; Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy.
| |
Collapse
|
27
|
Fry LE, Fahy E, Chrysostomou V, Hui F, Tang J, van Wijngaarden P, Petrou S, Crowston JG. The coma in glaucoma: Retinal ganglion cell dysfunction and recovery. Prog Retin Eye Res 2018; 65:77-92. [DOI: 10.1016/j.preteyeres.2018.04.001] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 03/18/2018] [Accepted: 04/03/2018] [Indexed: 01/07/2023]
|
28
|
Carelli V, La Morgia C, Ross-Cisneros FN, Sadun AA. Optic neuropathies: the tip of the neurodegeneration iceberg. Hum Mol Genet 2018; 26:R139-R150. [PMID: 28977448 PMCID: PMC5886475 DOI: 10.1093/hmg/ddx273] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 07/10/2017] [Indexed: 01/06/2023] Open
Abstract
The optic nerve and the cells that give origin to its 1.2 million axons, the retinal ganglion cells (RGCs), are particularly vulnerable to neurodegeneration related to mitochondrial dysfunction. Optic neuropathies may range from non-syndromic genetic entities, to rare syndromic multisystem diseases with optic atrophy such as mitochondrial encephalomyopathies, to age-related neurodegenerative diseases such as Alzheimer’s and Parkinson’s disease where optic nerve involvement has, until recently, been a relatively overlooked feature. New tools are available to thoroughly investigate optic nerve function, allowing unparalleled access to this part of the central nervous system. Understanding the molecular pathophysiology of RGC neurodegeneration and optic atrophy, is key to broadly understanding the pathogenesis of neurodegenerative disorders, for monitoring their progression in describing the natural history, and ultimately as outcome measures to evaluate therapies. In this review, the different layers, from molecular to anatomical, that may contribute to RGC neurodegeneration and optic atrophy are tackled in an integrated way, considering all relevant players. These include RGC dendrites, cell bodies and axons, the unmyelinated retinal nerve fiber layer and the myelinated post-laminar axons, as well as olygodendrocytes and astrocytes, looked for unconventional functions. Dysfunctional mitochondrial dynamics, transport, homeostatic control of mitobiogenesis and mitophagic removal, as well as specific propensity to apoptosis may target differently cell types and anatomical settings. Ultimately, we can envisage new investigative approaches and therapeutic options that will speed the early diagnosis of neurodegenerative diseases and their cure.
Collapse
Affiliation(s)
- Valerio Carelli
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Chiara La Morgia
- IRCCS Institute of Neurological Sciences of Bologna, Bellaria Hospital, Bologna, Italy.,Department of Biomedical and Neuromotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | | | - Alfredo A Sadun
- Doheny Eye Institute, Los Angeles, CA 90033, USA.,Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA 90095, USA
| |
Collapse
|
29
|
OPA1: How much do we know to approach therapy? Pharmacol Res 2018; 131:199-210. [PMID: 29454676 DOI: 10.1016/j.phrs.2018.02.018] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 02/12/2018] [Accepted: 02/12/2018] [Indexed: 01/01/2023]
Abstract
OPA1 is a GTPase that controls several functions, such as mitochondrial dynamics and energetics, mtDNA maintenance and cristae integrity. In the last years, there have been described other cellular pathways and mechanisms involving OPA1 directly or through its interaction. All this new information, by implementing our knowledge on OPA1 is instrumental to elucidating the pathogenic mechanisms of OPA1 mutations. Indeed, these are associated with dominant optic atrophy (DOA), one of the most common inherited optic neuropathies, and with an increasing number of heterogeneous neurodegenerative disorders. In this review, we overview all recent findings on OPA1 protein functions, on its dysfunction and related clinical phenotypes, focusing on the current therapeutic options and future perspectives to treat DOA and the other associated neurological disorders due to OPA1 mutations.
Collapse
|
30
|
Eight human OPA1 isoforms, long and short: What are they for? BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2018; 1859:263-269. [PMID: 29382469 DOI: 10.1016/j.bbabio.2018.01.005] [Citation(s) in RCA: 111] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 01/17/2018] [Accepted: 01/21/2018] [Indexed: 12/31/2022]
Abstract
OPA1 is a dynamin-related GTPase that controls mitochondrial dynamics, cristae integrity, energetics and mtDNA maintenance. The exceptional complexity of this protein is determined by the presence, in humans, of eight different isoforms that, in turn, are proteolytically cleaved into combinations of membrane-anchored long forms and soluble short forms. Recent advances highlight how each OPA1 isoform is able to fulfill "essential" mitochondrial functions, whereas only some variants carry out "specialized" features. Long forms determine fusion, long or short forms alone build cristae, whereas long and short forms together tune mitochondrial morphology. These findings offer novel challenging therapeutic potential to gene therapy.
Collapse
|
31
|
Ito YA, Di Polo A. Mitochondrial dynamics, transport, and quality control: A bottleneck for retinal ganglion cell viability in optic neuropathies. Mitochondrion 2017; 36:186-192. [PMID: 28866056 DOI: 10.1016/j.mito.2017.08.014] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/11/2017] [Accepted: 08/29/2017] [Indexed: 01/03/2023]
Abstract
Retinal ganglion cells, the neurons that selectively die in glaucoma and other optic neuropathies, are endowed with an exceedingly active metabolism and display a particular vulnerability to mitochondrial dysfunction. Mitochondria are exquisitely dynamic organelles that are continually responding to endogenous and environmental cues to readily meet the energy demand of neuronal networks. The highly orchestrated regulation of mitochondrial biogenesis, fusion, fission, transport and degradation is paramount for the maintenance of energy-expensive synapses at RGC dendrites and axon terminals geared for optimal neurotransmission. The present review focuses on the progress made to date on understanding the biology of mitochondrial dynamics and quality control and how dysregulation of these processes can profoundly affect retinal ganglion cell viability and function in optic nerve diseases.
Collapse
Affiliation(s)
- Yoko A Ito
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Université de Montréal, Montreal, Quebec H2X 1R9, Canada
| | - Adriana Di Polo
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Department of Neuroscience, Université de Montréal, Montreal, Quebec H2X 1R9, Canada.
| |
Collapse
|
32
|
Williams PA, Harder JM, Foxworth NE, Cardozo BH, Cochran KE, John SWM. Nicotinamide and WLD S Act Together to Prevent Neurodegeneration in Glaucoma. Front Neurosci 2017; 11:232. [PMID: 28487632 PMCID: PMC5403885 DOI: 10.3389/fnins.2017.00232] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/07/2017] [Indexed: 01/09/2023] Open
Abstract
Glaucoma is a complex neurodegenerative disease characterized by progressive visual dysfunction leading to vision loss. Retinal ganglion cells are the primary affected neuronal population, with a critical insult damaging their axons in the optic nerve head. This insult is typically secondary to harmfully high levels of intraocular pressure (IOP). We have previously determined that early mitochondrial abnormalities within retinal ganglion cells lead to neuronal dysfunction, with age-related declines in NAD (NAD+ and NADH) rendering retinal ganglion cell mitochondria vulnerable to IOP-dependent stresses. The Wallerian degeneration slow allele, WldS, decreases the vulnerability of retinal ganglion cells in eyes with elevated IOP, but the exact mechanism(s) of protection from glaucoma are not determined. Here, we demonstrate that WldS increases retinal NAD levels. Coupled with nicotinamide administration (an NAD precursor), it robustly protects from glaucomatous neurodegeneration in a mouse model of glaucoma (94% of eyes having no glaucoma, more than WldS or nicotinamide alone). Importantly, nicotinamide and WldS protect somal, synaptic, and axonal compartments, prevent loss of anterograde axoplasmic transport, and protect from visual dysfunction as assessed by pattern electroretinogram. Boosting NAD production generally benefits major compartments of retinal ganglion cells, and may be of value in other complex, age-related, axonopathies where multiple neuronal compartments are ultimately affected.
Collapse
Affiliation(s)
- Pete A Williams
- The Jackson Laboratory, Howard Hughes Medical InstituteBar Harbor, ME, USA
| | - Jeffrey M Harder
- The Jackson Laboratory, Howard Hughes Medical InstituteBar Harbor, ME, USA
| | - Nicole E Foxworth
- The Jackson Laboratory, Howard Hughes Medical InstituteBar Harbor, ME, USA
| | - Brynn H Cardozo
- The Jackson Laboratory, Howard Hughes Medical InstituteBar Harbor, ME, USA
| | - Kelly E Cochran
- The Jackson Laboratory, Howard Hughes Medical InstituteBar Harbor, ME, USA
| | - Simon W M John
- The Jackson Laboratory, Howard Hughes Medical InstituteBar Harbor, ME, USA.,Department of Ophthalmology, Tufts University of MedicineBoston, MA, USA
| |
Collapse
|
33
|
Morgan JE, Tribble J, Fergusson J, White N, Erchova I. The optical detection of retinal ganglion cell damage. Eye (Lond) 2017; 31:199-205. [PMID: 28060357 PMCID: PMC5306469 DOI: 10.1038/eye.2016.290] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Accepted: 11/08/2016] [Indexed: 11/08/2022] Open
Abstract
We provide an overview of developments in the use optical coherence tomography (OCT) imaging for the detection of retinal ganglion cell (RGC) damage in vivo that avoid use of any exogenous ligands to label cells. The method employs high-resolution OCT using broad spectral light sources to deliver axial resolution of under 5 μm. The resolution approximates that of cellular organelles, which undergo degenerative changes that progress to apoptosis as a result of axon damage. These degenerative changes are manifest as the loss of RGC dendrites and fragmentation of the subcellular network of organelles, in particular, the mitochondria that support dendritic structure. These changes can alter the light-scattering behavior of degenerating neurons. Using OCT imaging techniques to identify these signals in cultured neurons, we have demonstrated changes in cultured cells and in retinal explants. Pilot studies in human glaucoma suggest that similar changes are detectable in the clinical setting. High-resolution OCT can be used to detect optical scatter signals that derive from the RGC/inner plexiform layer and are associated with neuronal damage. These findings suggest that OCT instruments can be used to derive quantitative measurements of RGC damage. Critically, these signals can be detected at an early stage of RGC degeneration when cells could be protected or remodeled to support visual recovery.
Collapse
Affiliation(s)
- J E Morgan
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, UK
| | - J Tribble
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, UK
| | - J Fergusson
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, UK
| | - N White
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, UK
| | - I Erchova
- School of Optometry and Vision Sciences, Cardiff University, Maindy Road, Cardiff, UK
| |
Collapse
|
34
|
Pérez MJ, Vergara-Pulgar K, Jara C, Cabezas-Opazo F, Quintanilla RA. Caspase-Cleaved Tau Impairs Mitochondrial Dynamics in Alzheimer’s Disease. Mol Neurobiol 2017; 55:1004-1018. [DOI: 10.1007/s12035-017-0385-x] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 01/04/2017] [Indexed: 11/29/2022]
|
35
|
Maes ME, Schlamp CL, Nickells RW. BAX to basics: How the BCL2 gene family controls the death of retinal ganglion cells. Prog Retin Eye Res 2017; 57:1-25. [PMID: 28064040 DOI: 10.1016/j.preteyeres.2017.01.002] [Citation(s) in RCA: 130] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/22/2016] [Accepted: 01/03/2017] [Indexed: 12/19/2022]
Abstract
Retinal ganglion cell (RGC) death is the principal consequence of injury to the optic nerve. For several decades, we have understood that the RGC death process was executed by apoptosis, suggesting that there may be ways to therapeutically intervene in this cell death program and provide a more direct treatment to the cells and tissues affected in diseases like glaucoma. A major part of this endeavor has been to elucidate the molecular biological pathways active in RGCs from the point of axonal injury to the point of irreversible cell death. A major component of this process is the complex interaction of members of the BCL2 gene family. Three distinct family members of proteins orchestrate the most critical junction in the apoptotic program of RGCs, culminating in the activation of pro-apoptotic BAX. Once active, BAX causes irreparable damage to mitochondria, while precipitating downstream events that finish off a dying ganglion cell. This review is divided into two major parts. First, we summarize the extent of knowledge of how BCL2 gene family proteins interact to facilitate the activation and function of BAX. This area of investigation has rapidly changed over the last few years and has yielded a dramatically different mechanistic understanding of how the intrinsic apoptotic program is run in mammalian cells. Second, we provided a comprehensive analysis of nearly two decades of investigation of the role of BAX in the process of RGC death, much of which has provided many important insights into the overall pathophysiology of diseases like glaucoma.
Collapse
Affiliation(s)
- Margaret E Maes
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Cassandra L Schlamp
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA
| | - Robert W Nickells
- Department of Ophthalmology and Visual Sciences, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
36
|
Song L, Yu A, Murray K, Cortopassi G. Bipolar cell reduction precedes retinal ganglion neuron loss in a complex 1 knockout mouse model. Brain Res 2016; 1657:232-244. [PMID: 28027875 DOI: 10.1016/j.brainres.2016.12.019] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 12/19/2016] [Accepted: 12/20/2016] [Indexed: 01/03/2023]
Abstract
Inherited mitochondrial complex 1 deficiency causes Leber's hereditary Optic Neuropathy (LHON) and retinal ganglion cell (RGC) degeneration, and optic neuropathies are common in many inherited mitochondrial diseases. How mitochondrial defects pathomechanistically trigger optic neuropathy remains unclear. We observe that complex 1-deficient Ndufs4-/- mice present with acute vision loss around p30, and this vision loss is coincident with an 'inflammatory wave'. In order to understand what causes the inflammatory wave we explored retinal pathology that occurs from p20-p30. The results indicated that in the period p20-p30 in Ndufs4-/- retinas, there is: significant reduction in bipolar cells, RGC dendritic atrophy, reduced PSD95, increased oxidative stress as manifested by increased 4HNE, HO1 and Cuzn-SOD, increased mitochondrial biogenesis and increased apoptosis. These precede the major induction of 'inflammatory wave' at p30 shown previously, but occur earlier than frank RGC loss at p42. In general, complex 1 deficiency in retina triggers oxidative stress and mitochondrial respiratory dysfunction that causes death of the most sensitive cells, including bipolar cells and their synaptic contacts and amacrine cells in the early period, 20-24days. The early death of these cells is the likely precursor to the sharp rise in inflammatory molecules that occurs at day 30 and coincides with vision loss, and greatly precedes the death of RGCs that occurs at p42. These data suggest that metabolic antioxidant support of the most sensitive cells in the retina, or anti-inflammatory suppression of the consequences of their death, are both rational strategies for mitochondrial blinding disease.
Collapse
Affiliation(s)
- Lanying Song
- Vet Med: Molecular Biosciences, University of California, Davis, Davis, CA 95616, United States
| | - Alfred Yu
- Vet Med: Molecular Biosciences, University of California, Davis, Davis, CA 95616, United States
| | - Karl Murray
- Center for Neuroscience, University of California, Davis, Davis, CA 95616, United States; Department of Psychiatry & Behavioral Sciences, University of California, Davis, Davis, CA 95616, United States
| | - Gino Cortopassi
- Vet Med: Molecular Biosciences, University of California, Davis, Davis, CA 95616, United States.
| |
Collapse
|
37
|
Moulis MF, Millet AM, Daloyau M, Miquel MC, Ronsin B, Wissinger B, Arnauné-Pelloquin L, Belenguer P. OPA1 haploinsufficiency induces a BNIP3-dependent decrease in mitophagy in neurons: relevance to Dominant Optic Atrophy. J Neurochem 2016; 140:485-494. [DOI: 10.1111/jnc.13894] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 09/28/2016] [Accepted: 10/24/2016] [Indexed: 12/31/2022]
Affiliation(s)
- Manon F Moulis
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Aurélie M Millet
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Marlène Daloyau
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Marie-Christine Miquel
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Brice Ronsin
- Center of Developmental Biology (CBD); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Bernd Wissinger
- Center for Ophthalmology; University of Tübingen; Tübingen Germany
| | - Laetitia Arnauné-Pelloquin
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| | - Pascale Belenguer
- Research Center on Animal Cognition (CRCA); Center for Integrative Biology (CBI); Toulouse University; CNRS UPS France
| |
Collapse
|
38
|
A neurodegenerative perspective on mitochondrial optic neuropathies. Acta Neuropathol 2016; 132:789-806. [PMID: 27696015 PMCID: PMC5106504 DOI: 10.1007/s00401-016-1625-2] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2016] [Revised: 09/24/2016] [Accepted: 09/25/2016] [Indexed: 12/15/2022]
Abstract
Mitochondrial optic neuropathies constitute an important cause of chronic visual morbidity and registrable blindness in both the paediatric and adult population. It is a genetically heterogeneous group of disorders caused by both mitochondrial DNA (mtDNA) mutations and a growing list of nuclear genetic defects that invariably affect a critical component of the mitochondrial machinery. The two classical paradigms are Leber hereditary optic neuropathy (LHON), which is a primary mtDNA disorder, and autosomal dominant optic atrophy (DOA) secondary to pathogenic mutations within the nuclear gene OPA1 that encodes for a mitochondrial inner membrane protein. The defining neuropathological feature is the preferential loss of retinal ganglion cells (RGCs) within the inner retina but, rather strikingly, the smaller calibre RGCs that constitute the papillomacular bundle are particularly vulnerable, whereas melanopsin-containing RGCs are relatively spared. Although the majority of patients with LHON and DOA will present with isolated optic nerve involvement, some individuals will also develop additional neurological complications pointing towards a greater vulnerability of the central nervous system (CNS) in susceptible mutation carriers. These so-called “plus” phenotypes are mechanistically important as they put the loss of RGCs within the broader perspective of neuronal loss and mitochondrial dysfunction, highlighting common pathways that could be modulated to halt progressive neurodegeneration in other related CNS disorders. The management of patients with mitochondrial optic neuropathies still remains largely supportive, but the development of effective disease-modifying treatments is now within tantalising reach helped by major advances in drug discovery and delivery, and targeted genetic manipulation.
Collapse
|
39
|
Cai YL, Zou YC, Lei JH, Zeng GP, Wang Y. The investigation on the role of mitochondrial fusion protein 1 in the development of myopia. Indian J Ophthalmol 2016; 64:500-3. [PMID: 27609161 PMCID: PMC5026074 DOI: 10.4103/0301-4738.190137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Purpose: The aim of this study is to preliminarily investigate the expression of mitochondrial fusion protein 1 (MFN1) in a lens-induced animal myopia (LIM) model and to explore the relationship between MFN1 and the visual development. Materials and Methods: MFN1 gene expression in guinea pigs was examined during the development of minus LIM, 15 tri-colored guinea pigs were obtained, and one eye of each pig was randomly selected and treated with −7.00D lenses. Ocular refraction and axial length were collected before intervention and 1, 2, and 3 weeks after intervention. After the refraction and axial length measurements at 1, 2, and 3 weeks of lens intervention, five guinea pigs were randomly selected. MFN1 expression in the retina of both eyes was tested by immunohistochemistry technique. Results: MFN1-positive cells could be observed in the retina of both eyes. The positive cells in the LIM eyes were staining deeper, and much more positive cells could be observed. Furthermore, MFN1-positive expression could be seen mainly in ganglion cells after 1 week of minus lens intervention, and with time extension, more and more positive cells appeared in the rod-cone cell and bipolar cell layer, and this phenomenon could not be found in the normal control eyes. Conclusion: This study suggested that MFN1 might be correlated to the development of myopia.
Collapse
Affiliation(s)
- Yun-Lin Cai
- The Second Clinical Medical College of North Sichuan Medical College, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Yun-Chun Zou
- Department of Ophthalmology and Optometry, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jia-Hong Lei
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Guan-Peng Zeng
- Department of Ophthalmology and Optometry, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Ying Wang
- Department of Ophthalmology and Optometry, North Sichuan Medical College, Nanchong, Sichuan, China
| |
Collapse
|
40
|
Kushnareva Y, Seong Y, Andreyev AY, Kuwana T, Kiosses WB, Votruba M, Newmeyer DD. Mitochondrial dysfunction in an Opa1(Q285STOP) mouse model of dominant optic atrophy results from Opa1 haploinsufficiency. Cell Death Dis 2016; 7:e2309. [PMID: 27468686 PMCID: PMC4973340 DOI: 10.1038/cddis.2016.160] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 04/29/2016] [Accepted: 05/02/2016] [Indexed: 12/22/2022]
Abstract
Mutations in the opa1 (optic atrophy 1) gene lead to autosomal dominant optic atrophy (ADOA), a hereditary eye disease. This gene encodes the Opa1 protein, a mitochondrial dynamin-related GTPase required for mitochondrial fusion and the maintenance of normal crista structure. The majority of opa1 mutations encode truncated forms of the protein, lacking a complete GTPase domain. It is unclear whether the phenotype results from haploinsufficiency or rather a deleterious effect of truncated Opa1 protein. We studied a heterozygous Opa1 mutant mouse carrying a defective allele with a stop codon in the beginning of the GTPase domain at residue 285, a mutation that mimics human pathological mutations. Using an antibody raised against an N-terminal portion of Opa1, we found that the level of wild-type protein was decreased in the mutant mice, as predicted. However, no truncated Opa1 protein was expressed. In embryonic fibroblasts isolated from the mutant mice, this partial loss of Opa1 caused mitochondrial respiratory deficiency and a selective loss of respiratory Complex IV subunits. Furthermore, partial Opa1 deficiency resulted in a substantial resistance to endoplasmic reticulum stress-induced death. On the other hand, the enforced expression of truncated Opa1 protein in cells containing normal levels of wild-type protein did not cause mitochondrial defects. Moreover, cells expressing the truncated Opa1 protein showed reduced Bax activation in response to apoptotic stimuli. Taken together, our results exclude deleterious dominant-negative or gain-of-function mechanisms for this type of Opa1 mutation and affirm haploinsufficiency as the mechanism underlying mitochondrial dysfunction in ADOA.
Collapse
Affiliation(s)
- Y Kushnareva
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - Y Seong
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - A Y Andreyev
- Department of Pharmacology, University of California San Diego, La Jolla, CA 92093, USA
| | - T Kuwana
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - W B Kiosses
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | - M Votruba
- School of Optometry and Vision Sciences, Cardiff University, Cardiff CF24 4LU, UK.,Cardiff Eye Unit, University Hospital Wales, Cardiff CF14 4XW, UK
| | - D D Newmeyer
- Immune Regulation, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| |
Collapse
|
41
|
A randomized, placebo-controlled trial of the benzoquinone idebenone in a mouse model of OPA1-related dominant optic atrophy reveals a limited therapeutic effect on retinal ganglion cell dendropathy and visual function. Neuroscience 2016; 319:92-106. [PMID: 26820596 DOI: 10.1016/j.neuroscience.2016.01.042] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 12/19/2022]
Abstract
Dominant optic atrophy (DOA) arises from mutations in the OPA1 gene that promotes fusion of the inner mitochondrial membrane and plays a role in maintaining ATP levels. Patients display optic disc pallor, retinal ganglion cell (RGC) loss and bilaterally reduced vision. We report a randomized, placebo-controlled trial of idebenone at 2000 mg/kg/day in 56 Opa1 mutant mice (B6;C3-Opa1(Q285STOP)), with RGC dendropathy and visual loss, and 63 wildtype mice. We assessed cellular responses in the retina, brain and liver and RGC morphology, by diolistic labeling, Sholl analysis and quantification of dendritic morphometric features. Vision was assessed by optokinetic responses. ATP levels were raised by 0.57 nmol/mg (97.73%, p=0.035) in brain from idebenone-treated Opa1 mutant mice, but in the liver there was an 80.35% (p=0.011) increase in oxidative damage. NQO1 expression in Opa1 mutant mice was reduced in the brain (to 30.5%, p=0.002) but not in retina, and neither expression level was induced by idebenone. ON-center RGCs failed to show major recovery, other than improvements in secondary dendritic length (by 53.89%, p=0.052) and dendritic territory (by 2.22 × 10(4) μm(2) or 90.24%, p=0.074). An improvement in optokinetic response was observed (by 12.2 ± 3.2s, p=0.003), but this effect was not sustained over time. OFF-center RGCs from idebenone-treated wildtype mice showed shrinkage in total dendritic length by 2.40 mm (48.05%, p=0.025) and a 47.37% diminished Sholl profile (p=0.029). Visual function in wildtype idebenone-treated mice was impaired (2.9 fewer head turns than placebo, p=0.007). Idebenone appears largely ineffective in protecting Opa1 heterozygous RGCs from dendropathy. The detrimental effect of idebenone in wildtype mice has not been previously observed and raises some concerns.
Collapse
|
42
|
Rinholm JE, Vervaeke K, Tadross MR, Tkachuk AN, Kopek BG, Brown TA, Bergersen LH, Clayton DA. Movement and structure of mitochondria in oligodendrocytes and their myelin sheaths. Glia 2016; 64:810-25. [PMID: 26775288 DOI: 10.1002/glia.22965] [Citation(s) in RCA: 74] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Accepted: 12/22/2015] [Indexed: 12/30/2022]
Abstract
Mitochondria play several crucial roles in the life of oligodendrocytes. During development of the myelin sheath they are essential providers of carbon skeletons and energy for lipid synthesis. During normal brain function their consumption of pyruvate will be a key determinant of how much lactate is available for oligodendrocytes to export to power axonal function. Finally, during calcium-overload induced pathology, as occurs in ischemia, mitochondria may buffer calcium or induce apoptosis. Despite their important functions, very little is known of the properties of oligodendrocyte mitochondria, and mitochondria have never been observed in the myelin sheaths. We have now used targeted expression of fluorescent mitochondrial markers to characterize the location and movement of mitochondria within oligodendrocytes. We show for the first time that mitochondria are able to enter and move within the myelin sheath. Within the myelin sheath the highest number of mitochondria was in the cytoplasmic ridges along the sheath. Mitochondria moved more slowly than in neurons and, in contrast to their behavior in neurons and astrocytes, their movement was increased rather than inhibited by glutamate activating NMDA receptors. By electron microscopy we show that myelin sheath mitochondria have a low surface area of cristae, which suggests a low ATP production. These data specify fundamental properties of the oxidative phosphorylation system in oligodendrocytes, the glial cells that enhance cognition by speeding action potential propagation and provide metabolic support to axons.
Collapse
Affiliation(s)
- Johanne E Rinholm
- Department of Anatomy, The Brain and Muscle Energy Group, University of Oslo, Oslo, Norway.,Department of Oral Biology, The Brain and Muscle Energy Group, Electron Microscopic Laboratory, University of Oslo, Oslo, Norway.,Howard Hughes Medical Institute, Ashburn, Virginia
| | - Koen Vervaeke
- Howard Hughes Medical Institute, Ashburn, Virginia.,Department of Physiology, Laboratory of Neural Computation, University of Oslo, Oslo, Norway
| | | | | | | | | | - Linda H Bergersen
- Department of Oral Biology, The Brain and Muscle Energy Group, Electron Microscopic Laboratory, University of Oslo, Oslo, Norway
| | | |
Collapse
|
43
|
Faits MC, Zhang C, Soto F, Kerschensteiner D. Dendritic mitochondria reach stable positions during circuit development. eLife 2016; 5:e11583. [PMID: 26742087 PMCID: PMC4749546 DOI: 10.7554/elife.11583] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2015] [Accepted: 12/30/2015] [Indexed: 01/07/2023] Open
Abstract
Mitochondria move throughout neuronal dendrites and localize to sites of energy demand. The prevailing view of dendritic mitochondria as highly motile organelles whose distribution is continually adjusted by neuronal activity via Ca(2+)-dependent arrests is based on observations in cultured neurons exposed to artificial stimuli. Here, we analyze the movements of mitochondria in ganglion cell dendrites in the intact retina. We find that whereas during development 30% of mitochondria are motile at any time, as dendrites mature, mitochondria all but stop moving and localize stably to synapses and branch points. Neither spontaneous nor sensory-evoked activity and Ca(2+) transients alter motility of dendritic mitochondria; and pathological hyperactivity in a mouse model of retinal degeneration elevates rather than reduces motility. Thus, our findings indicate that dendritic mitochondria reach stable positions during a critical developmental period of high motility, and challenge current views about the role of activity in regulating mitochondrial transport in dendrites.
Collapse
Affiliation(s)
- Michelle C Faits
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, United States.,Graduate Program in Developmental, Regenerative and Stem Cell Biology, Washington University School of Medicine, St. Louis, United States
| | - Chunmeng Zhang
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, United States
| | - Florentina Soto
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, United States
| | - Daniel Kerschensteiner
- Department of Ophthalmology and Visual Sciences, Washington University School of Medicine, Saint Louis, United States.,Department of Neuroscience, Washington University School of Medicine, Saint Louis, United States.,Department of Biomedical Engineering, Washington University School of Medicine, Saint Louis, United States.,Hope Center for Neurological Disorders, Washington University School of Medicine, Saint Louis, United States
| |
Collapse
|
44
|
Abstract
Mitochondria are an essential component of multicellular life - from primitive organisms, to highly complex entities like mammals. The importance of mitochondria is underlined by their plethora of well-characterized essential functions such as energy production through oxidative phosphorylation (OX-PHOS), calcium and reactive oxygen species (ROS) signaling, and regulation of apoptosis. In addition, novel roles and attributes of mitochondria are coming into focus through the recent years of mitochondrial research. In particular, over the past decade the study of mitochondrial shape and dynamics has achieved special significance, as they are found to impact mitochondrial function. Recent advances indicate that mitochondrial function and dynamics are inter-connected, and maintain the balance between health and disease at a cellular and an organismal level. For example, excessive mitochondrial division (fission) is associated with functional defects, and is implicated in multiple human diseases from neurodegenerative diseases to cancer. In this chapter we examine the recent literature on the mitochondrial dynamics-function relationship, and explore how it impacts on the development and progression of human diseases. We will also highlight the implications of therapeutic manipulation of mitochondrial dynamics in treating various human pathologies.
Collapse
|
45
|
Zou YC, Lei JH, Wang Y, Xu S. Correlation between polymorphisms in the MFN1 gene and myopia in Chinese population. Int J Ophthalmol 2015; 8:1126-30. [PMID: 26682159 DOI: 10.3980/j.issn.2222-3959.2015.06.08] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 04/07/2015] [Indexed: 11/02/2022] Open
Abstract
AIM To explore whether genetic variations in the MFN1 gene are associated with low to moderate myopia in Chinese population. METHODS The case-control association analysis was used. The study included 100 independent myopia patients (-0.75 D ≤ spherical refraction ≤-8.00 D) and 100 sex-matched healthy controls (with binocular spherical equivalent ranges between -0.50 D and +0.50 D). Four single nucleotide polymorphism (SNP) tags (rs3976523, rs13098637, rs6762399 and rs7618348) were selected for genotyping by direct sequencing. The frequencies of genotypes and their alleles were calculated based on the number of SNP genotypes in each sample. The Chi-square test was used to examine the difference in the frequency between the myopia cases and controls. RESULTS Genotype distributions in the four SNPs were all in accordance with the Hardy-Weinberg equilibrium; analysis showed that rs13098637 was significantly associated with low to moderate myopia (P=0.003 and empirical P=0.010). There were no statistically significant differences observed for the genotype or allele frequencies of the other three SNPs between the myopia cases and controls in the Chinese population in this study. CONCLUSION The current study has revealed that the C allele of rs13098637 in MFN1 had a significant association with low to moderate myopia.
Collapse
Affiliation(s)
- Yun-Chun Zou
- Department of Ophthalmology and Optometry, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Jia-Hong Lei
- Department of Laboratory Medicine, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Ying Wang
- Department of Ophthalmology and Optometry, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| | - Shuang Xu
- Department of Ophthalmology and Optometry, North Sichuan Medical College, Nanchong 637000, Sichuan Province, China
| |
Collapse
|
46
|
Integrative properties of retinal ganglion cell electrical responsiveness depend on neurotrophic support and genotype in the mouse. Exp Eye Res 2015; 145:68-74. [PMID: 26614910 DOI: 10.1016/j.exer.2015.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2015] [Revised: 10/30/2015] [Accepted: 11/09/2015] [Indexed: 12/18/2022]
Abstract
Early stages of glaucoma and optic neuropathies are thought to show inner retina remodeling and functional changes of retinal ganglion cells (RGCs) before they die. To assess RGC functional plasticity, we investigated the contrast-gain control properties of the pattern electroretinogram (PERG), a sensitive measure of RGC function, as an index of spatio-temporal integration occurring in the inner retina circuitry subserving PERG generators. We studied the integrative properties of the PERG in mice exposed to different conditions of neurotrophic support. We also investigated the effect of genotypic differences among mouse strains with different susceptibility to glaucoma (C57BL/6J, DBA/2J, DBA/2.Gpnmb(+)). Results show that the integrative properties of the PERG recorded in the standard C57BL/6J inbred mouse strain are impaired after deficit of neurotrophic support and partially restored after exogenous neurotrophic administration. Changes in PERG amplitude, latency, and contrast-dependent responses differ between mouse strains with different susceptibility to glaucoma. Results represent a proof of concept that the PERG could be used as a tool for in-vivo monitoring of RGC functional plasticity before RGC death, the effect of neuroactive treatments, as well as for high-throughput tool for phenotypic screening of different mouse genotypes.
Collapse
|
47
|
Angebault C, Guichet PO, Talmat-Amar Y, Charif M, Gerber S, Fares-Taie L, Gueguen N, Halloy F, Moore D, Amati-Bonneau P, Manes G, Hebrard M, Bocquet B, Quiles M, Piro-Mégy C, Teigell M, Delettre C, Rossel M, Meunier I, Preising M, Lorenz B, Carelli V, Chinnery PF, Yu-Wai-Man P, Kaplan J, Roubertie A, Barakat A, Bonneau D, Reynier P, Rozet JM, Bomont P, Hamel CP, Lenaers G. Recessive Mutations in RTN4IP1 Cause Isolated and Syndromic Optic Neuropathies. Am J Hum Genet 2015; 97:754-60. [PMID: 26593267 DOI: 10.1016/j.ajhg.2015.09.012] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 09/25/2015] [Indexed: 12/28/2022] Open
Abstract
Autosomal-recessive optic neuropathies are rare blinding conditions related to retinal ganglion cell (RGC) and optic-nerve degeneration, for which only mutations in TMEM126A and ACO2 are known. In four families with early-onset recessive optic neuropathy, we identified mutations in RTN4IP1, which encodes a mitochondrial ubiquinol oxydo-reductase. RTN4IP1 is a partner of RTN4 (also known as NOGO), and its ortholog Rad8 in C. elegans is involved in UV light response. Analysis of fibroblasts from affected individuals with a RTN4IP1 mutation showed loss of the altered protein, a deficit of mitochondrial respiratory complex I and IV activities, and increased susceptibility to UV light. Silencing of RTN4IP1 altered the number and morphogenesis of mouse RGC dendrites in vitro and the eye size, neuro-retinal development, and swimming behavior in zebrafish in vivo. Altogether, these data point to a pathophysiological mechanism responsible for RGC early degeneration and optic neuropathy and linking RTN4IP1 functions to mitochondrial physiology, response to UV light, and dendrite growth during eye maturation.
Collapse
Affiliation(s)
- Claire Angebault
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Pierre-Olivier Guichet
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Yasmina Talmat-Amar
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Majida Charif
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France; INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France
| | - Sylvie Gerber
- INSERM U1163, Hôpital Necker Enfants-Malades, 75015 Paris, France
| | - Lucas Fares-Taie
- INSERM U1163, Hôpital Necker Enfants-Malades, 75015 Paris, France
| | - Naig Gueguen
- INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France
| | - François Halloy
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - David Moore
- Institute of Genetic Medicine, Centre for Life, Newcastle University and Wellcome Trust Centre for Mitochondrial Research, NE1 3BZ Newcastle upon Tyne, UK
| | - Patrizia Amati-Bonneau
- INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France
| | - Gael Manes
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Maxime Hebrard
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Béatrice Bocquet
- Centre de Référence pour les Maladies Sensorielles Génétiques, Hôpital Gui de Chauliac, CHRU Montpellier, 34090 Montpellier, France
| | - Mélanie Quiles
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Camille Piro-Mégy
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Marisa Teigell
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Cécile Delettre
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Mireille Rossel
- INSERM U710, Laboratoire MMDN EPHE, 34090 Montpellier, France
| | - Isabelle Meunier
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France; Centre de Référence pour les Maladies Sensorielles Génétiques, Hôpital Gui de Chauliac, CHRU Montpellier, 34090 Montpellier, France
| | - Markus Preising
- Department of Ophthalmology, Justus-Liebig University, 35392 Giessen, Germany
| | - Birgit Lorenz
- Department of Ophthalmology, Justus-Liebig University, 35392 Giessen, Germany
| | - Valerio Carelli
- IRCCS, Institute of Neurological Sciences of Bologna, Bellaria Hospital, 40139 Bologna, Italy; Department of Biomedical and NeuroMotor Sciences, University of Bologna, 40139 Bologna, Italy
| | - Patrick F Chinnery
- Institute of Genetic Medicine, Centre for Life, Newcastle University and Wellcome Trust Centre for Mitochondrial Research, NE1 3BZ Newcastle upon Tyne, UK
| | - Patrick Yu-Wai-Man
- Institute of Genetic Medicine, Centre for Life, Newcastle University and Wellcome Trust Centre for Mitochondrial Research, NE1 3BZ Newcastle upon Tyne, UK; Newcastle Eye Centre, Royal Victoria Infirmary, NE1 4LP Newcastle upon Tyne, UK
| | - Josseline Kaplan
- INSERM U1163, Hôpital Necker Enfants-Malades, 75015 Paris, France
| | - Agathe Roubertie
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France; Centre de Référence pour les Maladies Sensorielles Génétiques, Hôpital Gui de Chauliac, CHRU Montpellier, 34090 Montpellier, France
| | - Abdelhamid Barakat
- Laboratoire de Génétique Moléculaire Humaine, Département de Recherche Scientifique, Institut Pasteur du Maroc, 20360 Casablanca, Morocco
| | - Dominique Bonneau
- INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France
| | - Pascal Reynier
- INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France
| | | | - Pascale Bomont
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France
| | - Christian P Hamel
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France; Centre de Référence pour les Maladies Sensorielles Génétiques, Hôpital Gui de Chauliac, CHRU Montpellier, 34090 Montpellier, France
| | - Guy Lenaers
- INSERM U1051, Institut des Neurosciences de Montpellier, Université de Montpellier, 34090 Montpellier, France; INSERM U1083, CNRS 6214, Département de Biochimie et Génétique, Université LUNAM and Centre Hospitalier Universitaire, 49933 Angers, France.
| |
Collapse
|
48
|
Bertholet AM, Delerue T, Millet AM, Moulis MF, David C, Daloyau M, Arnauné-Pelloquin L, Davezac N, Mils V, Miquel MC, Rojo M, Belenguer P. Mitochondrial fusion/fission dynamics in neurodegeneration and neuronal plasticity. Neurobiol Dis 2015; 90:3-19. [PMID: 26494254 DOI: 10.1016/j.nbd.2015.10.011] [Citation(s) in RCA: 255] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 09/16/2015] [Accepted: 10/13/2015] [Indexed: 12/17/2022] Open
Abstract
Mitochondria are dynamic organelles that continually move, fuse and divide. The dynamic balance of fusion and fission of mitochondria determines their morphology and allows their immediate adaptation to energetic needs, keeps mitochondria in good health by restoring or removing damaged organelles or precipitates cells in apoptosis in cases of severe defects. Mitochondrial fusion and fission are essential in mammals and their disturbances are associated with several diseases. However, while mitochondrial fusion/fission dynamics, and the proteins that control these processes, are ubiquitous, associated diseases are primarily neurological disorders. Accordingly, inactivation of the main actors of mitochondrial fusion/fission dynamics is associated with defects in neuronal development, plasticity and functioning, both ex vivo and in vivo. Here, we present the central actors of mitochondrial fusion and fission and review the role of mitochondrial dynamics in neuronal physiology and pathophysiology. Particular emphasis is placed on the three main actors of these processes i.e. DRP1,MFN1-2, and OPA1 as well as on GDAP1, a protein of the mitochondrial outer membrane preferentially expressed in neurons. This article is part of a Special Issue entitled: Mitochondria & Brain.
Collapse
Affiliation(s)
- A M Bertholet
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - T Delerue
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - A M Millet
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - M F Moulis
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - C David
- CNRS, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France
| | - M Daloyau
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - L Arnauné-Pelloquin
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - N Davezac
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - V Mils
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - M C Miquel
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France
| | - M Rojo
- CNRS, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France; Université de Bordeaux, Institut de Biochimie et Génétique Cellulaires (IBGC), UMR5095, Bordeaux, France.
| | - P Belenguer
- Université de Toulouse, Centre de Biologie du Développement, CNRS, UMR5547/Université Paul Sabatier, Toulouse, France; CNRS, Centre de Biologie du Développement, UMR5547/Université Paul Sabatier, Toulouse, France.
| |
Collapse
|
49
|
Chrysostomou A, Grady JP, Laude A, Taylor RW, Turnbull DM, Lax NZ. Investigating complex I deficiency in Purkinje cells and synapses in patients with mitochondrial disease. Neuropathol Appl Neurobiol 2015; 42:477-92. [PMID: 26337858 PMCID: PMC4973693 DOI: 10.1111/nan.12282] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 08/20/2015] [Indexed: 12/21/2022]
Abstract
Aims Cerebellar ataxia is common in patients with mitochondrial disease, and despite previous neuropathological investigations demonstrating vulnerability of the olivocerebellar pathway in patients with mitochondrial disease, the exact neurodegenerative mechanisms are still not clear. We use quantitative quadruple immunofluorescence to enable precise quantification of mitochondrial respiratory chain protein expression in Purkinje cell bodies and their synaptic terminals in the dentate nucleus. Methods We investigated NADH dehydrogenase [ubiquinone] 1 alpha subcomplex subunit 13 protein expression in 12 clinically and genetically defined patients with mitochondrial disease and ataxia and 10 age‐matched controls. Molecular genetic analysis was performed to determine heteroplasmy levels of mutated mitochondrial DNA in Purkinje cell bodies and inhibitory synapses. Results Our data reveal that complex I deficiency is present in both Purkinje cell bodies and their inhibitory synapses which surround dentate nucleus neurons. Inhibitory synapses are fewer and enlarged in patients which could represent a compensatory mechanism. Mitochondrial DNA heteroplasmy demonstrated similarly high levels of mutated mitochondrial DNA in cell bodies and synapses. Conclusions This is the first study to use a validated quantitative immunofluorescence technique to determine complex I expression in neurons and presynaptic terminals, evaluating the distribution of respiratory chain deficiencies and assessing the degree of morphological abnormalities affecting synapses. Respiratory chain deficiencies detected in Purkinje cell bodies and their synapses and structural synaptic changes are likely to contribute to altered cerebellar circuitry and progression of ataxia.
Collapse
Affiliation(s)
- Alexia Chrysostomou
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - John P Grady
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Alex Laude
- Bio-imaging Unit, Newcastle University, Newcastle upon Tyne, UK
| | - Robert W Taylor
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Doug M Turnbull
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| | - Nichola Z Lax
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
50
|
Abstract
Mitochondrial dysfunction underlies many human disorders, including those that affect the visual system. The retinal ganglion cells, whose axons form the optic nerve, are often damaged by mitochondrial-related diseases which result in blindness. Both mitochondrial DNA (mtDNA) and nuclear gene mutations impacting many different mitochondrial processes can result in optic nerve disease. Of particular importance are mutations that impair mitochondrial network dynamics (fusion and fission), oxidative phosphorylation (OXPHOS), and formation of iron-sulfur complexes. Current genetic knowledge can inform genetic counseling and suggest strategies for novel gene-based therapies. Identifying new optic neuropathy-causing genes and defining the role of current and novel genes in disease will be important steps toward the development of effective and potentially neuroprotective therapies.
Collapse
Affiliation(s)
- Janey L Wiggs
- Department of Ophthalmology, Harvard Medical School and Massachusetts Eye and Ear, Boston, Massachusetts 02114;
| |
Collapse
|