1
|
Klimenko ES, Sukhareva KS, Vlasova Y, Smolina NA, Fomicheva Y, Knyazeva A, Muravyev AS, Sorokina MY, Gavrilova LS, Boldyreva LV, Medvedeva SS, Sejersen T, Kostareva AA. Flnc expression impacts mitochondrial function, autophagy, and calcium handling in C2C12 cells. Exp Cell Res 2024; 442:114174. [PMID: 39089502 DOI: 10.1016/j.yexcr.2024.114174] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/12/2024] [Accepted: 07/21/2024] [Indexed: 08/04/2024]
Affiliation(s)
- E S Klimenko
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - K S Sukhareva
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - YuA Vlasova
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - N A Smolina
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - YuV Fomicheva
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - A Knyazeva
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - A S Muravyev
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - M Yu Sorokina
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - L S Gavrilova
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - L V Boldyreva
- Scientific-Research Institute of Neurosciences and Medicine, Novosibirsk, Russia; Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - S S Medvedeva
- Scientific-Research Institute of Neurosciences and Medicine, Novosibirsk, Russia; Institute of Molecular and Cellular Biology, Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - T Sejersen
- Department of Women's and Children's Health, Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong
| | - A A Kostareva
- Almazov National Medical Research Centre, Saint-Petersburg, Russia; Department of Women's and Children's Health, Karolinska Institutet, Department of Child Neurology, Karolinska University Hospital, Astrid Lindgren Children's Hospital, Stockholm, Sweden; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong.
| |
Collapse
|
2
|
Mak G, Tarnopolsky M, Lu JQ. Secondary mitochondrial dysfunction across the spectrum of hereditary and acquired muscle disorders. Mitochondrion 2024; 78:101945. [PMID: 39134108 DOI: 10.1016/j.mito.2024.101945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Revised: 07/15/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024]
Abstract
Mitochondria form a dynamic network within skeletal muscle. This network is not only responsible for producing adenosine triphosphate (ATP) through oxidative phosphorylation, but also responds through fission, fusion and mitophagy to various factors, such as increased energy demands, oxidative stress, inflammation, and calcium dysregulation. Mitochondrial dysfunction in skeletal muscle not only occurs in primary mitochondrial myopathies, but also other hereditary and acquired myopathies. As such, this review attempts to highlight the clinical and histopathologic aspects of mitochondrial dysfunction seen in hereditary and acquired myopathies, as well as discuss potential mechanisms leading to mitochondrial dysfunction and therapies to restore mitochondrial function.
Collapse
Affiliation(s)
- Gloria Mak
- University of Alberta, Department of Neurology, Edmonton, Alberta, Canada
| | - Mark Tarnopolsky
- McMaster University, Department of Medicine and Pediatrics, Hamilton, Ontario, Canada
| | - Jian-Qiang Lu
- McMaster University, Department of Pathology and Molecular Medicine, Hamilton, Ontario, Canada.
| |
Collapse
|
3
|
Berwanger C, Terres D, Pesta D, Eggers B, Marcus K, Wittig I, Wiesner RJ, Schröder R, Clemen CS. Immortalised murine R349P desmin knock-in myotubes exhibit a reduced proton leak and decreased ADP/ATP translocase levels in purified mitochondria. Eur J Cell Biol 2024; 103:151399. [PMID: 38412640 DOI: 10.1016/j.ejcb.2024.151399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 02/18/2024] [Accepted: 02/19/2024] [Indexed: 02/29/2024] Open
Abstract
Desmin gene mutations cause myopathies and cardiomyopathies. Our previously characterised R349P desminopathy mice, which carry the ortholog of the common human desmin mutation R350P, showed marked alterations in mitochondrial morphology and function in muscle tissue. By isolating skeletal muscle myoblasts from offspring of R349P desminopathy and p53 knock-out mice, we established an immortalised cellular disease model. Heterozygous and homozygous R349P desmin knock-in and wild-type myoblasts could be well differentiated into multinucleated spontaneously contracting myotubes. The desminopathy myoblasts showed the characteristic disruption of the desmin cytoskeleton and desmin protein aggregation, and the desminopathy myotubes showed the characteristic myofibrillar irregularities. Long-term electrical pulse stimulation promoted myotube differentiation and markedly increased their spontaneous contraction rate. In both heterozygous and homozygous R349P desminopathy myotubes, this treatment restored a regular myofibrillar cross-striation pattern as seen in wild-type myotubes. High-resolution respirometry of mitochondria purified from myotubes by density gradient ultracentrifugation revealed normal oxidative phosphorylation capacity, but a significantly reduced proton leak in mitochondria from the homozygous R349P desmin knock-in cells. Consistent with a reduced proton flux across the inner mitochondrial membrane, our quantitative proteomic analysis of the purified mitochondria revealed significantly reduced levels of ADP/ATP translocases in the homozygous R349P desmin knock-in genotype. As this alteration was also detected in the soleus muscle of R349P desminopathy mice, which, in contrast to the mitochondria purified from cultured cells, showed a variety of other dysregulated mitochondrial proteins, we consider this finding to be an early step in the pathogenesis of secondary mitochondriopathy in desminopathy.
Collapse
Affiliation(s)
- Carolin Berwanger
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Dominic Terres
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany
| | - Dominik Pesta
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), Medical Faculty, and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Britta Eggers
- Medizinisches Proteom-Center, Medical Faculty, and Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Medical Faculty, and Medical Proteome Analysis, Center for Proteindiagnostics (PRODI), Ruhr-University Bochum, Bochum, Germany
| | - Ilka Wittig
- Functional Proteomics, Institute for Cardiovascular Physiology, Goethe University, Frankfurt, Germany
| | - Rudolf J Wiesner
- Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany
| | - Rolf Schröder
- Department of Neuropathology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Christoph S Clemen
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Cologne, Germany; Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Cologne, Germany.
| |
Collapse
|
4
|
Hovhannisyan Y, Li Z, Callon D, Suspène R, Batoumeni V, Canette A, Blanc J, Hocini H, Lefebvre C, El-Jahrani N, Kitsara M, L'honoré A, Kordeli E, Fornes P, Concordet JP, Tachdjian G, Rodriguez AM, Vartanian JP, Béhin A, Wahbi K, Joanne P, Agbulut O. Critical contribution of mitochondria in the development of cardiomyopathy linked to desmin mutation. Stem Cell Res Ther 2024; 15:10. [PMID: 38167524 PMCID: PMC10763022 DOI: 10.1186/s13287-023-03619-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 12/18/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Beyond the observed alterations in cellular structure and mitochondria, the mechanisms linking rare genetic mutations to the development of heart failure in patients affected by desmin mutations remain unclear due in part, to the lack of relevant human cardiomyocyte models. METHODS To shed light on the role of mitochondria in these mechanisms, we investigated cardiomyocytes derived from human induced pluripotent stem cells carrying the heterozygous DESE439K mutation that were either isolated from a patient or generated by gene editing. To increase physiological relevance, cardiomyocytes were either cultured on an anisotropic micropatterned surface to obtain elongated and aligned cardiomyocytes, or as a cardiac spheroid to create a micro-tissue. Moreover, when applicable, results from cardiomyocytes were confirmed with heart biopsies of suddenly died patient of the same family harboring DESE439K mutation, and post-mortem heart samples from five control healthy donors. RESULTS The heterozygous DESE439K mutation leads to dramatic changes in the overall cytoarchitecture of cardiomyocytes, including cell size and morphology. Most importantly, mutant cardiomyocytes display altered mitochondrial architecture, mitochondrial respiratory capacity and metabolic activity reminiscent of defects observed in patient's heart tissue. Finally, to challenge the pathological mechanism, we transferred normal mitochondria inside the mutant cardiomyocytes and demonstrated that this treatment was able to restore mitochondrial and contractile functions of cardiomyocytes. CONCLUSIONS This work highlights the deleterious effects of DESE439K mutation, demonstrates the crucial role of mitochondrial abnormalities in the pathophysiology of desmin-related cardiomyopathy, and opens up new potential therapeutic perspectives for this disease.
Collapse
Affiliation(s)
- Yeranuhi Hovhannisyan
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France
| | - Zhenlin Li
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France
| | - Domitille Callon
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France
- Department of Pathology, Academic Hospital of Reims, Reims, France
| | - Rodolphe Suspène
- Virus and Cellular Stress Unit, Department of Virology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Vivien Batoumeni
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France
- Ksilink, Strasbourg, France
| | - Alexis Canette
- Service de Microscopie Électronique (IBPS-SME), Institut de Biologie Paris-Seine (IBPS), CNRS, Sorbonne Université, Paris, France
| | - Jocelyne Blanc
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France
| | - Hakim Hocini
- INSERM U955, Equipe 16, Université Paris-Est Créteil, Créteil, France
| | - Cécile Lefebvre
- INSERM U955, Equipe 16, Université Paris-Est Créteil, Créteil, France
| | - Nora El-Jahrani
- INSERM U955, Equipe 16, Université Paris-Est Créteil, Créteil, France
| | - Maria Kitsara
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France
| | - Aurore L'honoré
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France
| | - Ekaterini Kordeli
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France
| | - Paul Fornes
- Department of Pathology, Academic Hospital of Reims, Reims, France
| | - Jean-Paul Concordet
- INSERM U1154, CNRS UMR7196, Museum National d'Histoire Naturelle, Paris, France
| | - Gérard Tachdjian
- Laboratoire de Cytogénétique, Service d'Histologie-Embryologie-Cytogénétique, AP-HP, Hôpital Antoine Béclère, Université Paris Saclay, Clamart, France
| | - Anne-Marie Rodriguez
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France
| | - Jean-Pierre Vartanian
- Virus and Cellular Stress Unit, Department of Virology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Anthony Béhin
- Reference Center for Muscle Diseases Paris-Est, Myology Institute, AP-HP, Pitié-Salpêtrière Hospital, Sorbonne Université, Paris, France
| | - Karim Wahbi
- Cardiology Department, AP-HP, Cochin Hospital, Université Paris Cité, Paris, France
| | - Pierre Joanne
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France.
| | - Onnik Agbulut
- UMR CNRS 8256, INSERM U1164, Biological Adaptation and Ageing, Institut de Biologie Paris-Seine (IBPS), Sorbonne Université, 7, Quai St Bernard (case 256), 75005, Paris, France.
| |
Collapse
|
5
|
Pizzamiglio C, Hanna MG, Pitceathly RDS. Primary mitochondrial diseases. HANDBOOK OF CLINICAL NEUROLOGY 2024; 204:53-76. [PMID: 39322395 DOI: 10.1016/b978-0-323-99209-1.00004-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Primary mitochondrial diseases (PMDs) are a heterogeneous group of hereditary disorders characterized by an impairment of the mitochondrial respiratory chain. They are the most common group of genetic metabolic disorders, with a prevalence of 1 in 4,300 people. The presence of leukoencephalopathy is recognized as an important feature in many PMDs and can be a manifestation of mutations in both mitochondrial DNA (classic syndromes such as mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes; myoclonic epilepsy with ragged-red fibers [RRFs]; Leigh syndrome; and Kearns-Sayre syndrome) and nuclear DNA (mutations in maintenance genes such as POLG, MPV17, and TYMP; Leigh syndrome; and mitochondrial aminoacyl-tRNA synthetase disorders). In this chapter, PMDs associated with white matter involvement are outlined, including details of clinical presentations, brain MRI features, and elements of differential diagnoses. The current approach to the diagnosis of PMDs and management strategies are also discussed. A PMD diagnosis in a subject with leukoencephalopathy should be considered in the presence of specific brain MRI features (for example, cyst-like lesions, bilateral basal ganglia lesions, and involvement of both cerebral hemispheres and cerebellum), in addition to a complex neurologic or multisystem disorder. Establishing a genetic diagnosis is crucial to ensure appropriate genetic counseling, multidisciplinary team input, and eligibility for clinical trials.
Collapse
Affiliation(s)
- Chiara Pizzamiglio
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Michael G Hanna
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, United Kingdom
| | - Robert D S Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, United Kingdom; NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, National Hospital for Neurology and Neurosurgery, London, United Kingdom.
| |
Collapse
|
6
|
Lew SY, Mohd Hisam NS, Phang MWL, Syed Abdul Rahman SN, Poh RYY, Lim SH, Kamaruzzaman MA, Chau SC, Tsui KC, Lim LW, Wong KH. Adenosine Improves Mitochondrial Function and Biogenesis in Friedreich's Ataxia Fibroblasts Following L-Buthionine Sulfoximine-Induced Oxidative Stress. BIOLOGY 2023; 12:biology12040559. [PMID: 37106759 PMCID: PMC10136261 DOI: 10.3390/biology12040559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/25/2023] [Accepted: 03/30/2023] [Indexed: 04/29/2023]
Abstract
Adenosine is a nucleoside that is widely distributed in the central nervous system and acts as a central excitatory and inhibitory neurotransmitter in the brain. The protective role of adenosine in different pathological conditions and neurodegenerative diseases is mainly mediated by adenosine receptors. However, its potential role in mitigating the deleterious effects of oxidative stress in Friedreich's ataxia (FRDA) remains poorly understood. We aimed to investigate the protective effects of adenosine against mitochondrial dysfunction and impaired mitochondrial biogenesis in L-buthionine sulfoximine (BSO)-induced oxidative stress in dermal fibroblasts derived from an FRDA patient. The FRDA fibroblasts were pre-treated with adenosine for 2 h, followed by 12.50 mM BSO to induce oxidative stress. Cells in medium without any treatments or pre-treated with 5 µM idebenone served as the negative and positive controls, respectively. Cell viability, mitochondrial membrane potential (MMP), aconitase activity, adenosine triphosphate (ATP) level, mitochondrial biogenesis, and associated gene expressions were assessed. We observed disruption of mitochondrial function and biogenesis and alteration in gene expression patterns in BSO-treated FRDA fibroblasts. Pre-treatment with adenosine ranging from 0-600 µM restored MMP, promoted ATP production and mitochondrial biogenesis, and modulated the expression of key metabolic genes, namely nuclear respiratory factor 1 (NRF1), transcription factor A, mitochondrial (TFAM), and NFE2-like bZIP transcription factor 2 (NFE2L2). Our study demonstrated that adenosine targeted mitochondrial defects in FRDA, contributing to improved mitochondrial function and biogenesis, leading to cellular iron homeostasis. Therefore, we suggest a possible therapeutic role for adenosine in FRDA.
Collapse
Affiliation(s)
- Sze Yuen Lew
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | | | - Michael Weng Lok Phang
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | | | - Rozaida Yuen Ying Poh
- Department of Biomedical Science, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Siew Huah Lim
- Department of Chemistry, Faculty of Science, Universiti Malaya, Kuala Lumpur 50603, Malaysia
| | - Mohd Amir Kamaruzzaman
- Department of Anatomy, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latiff, Cheras, Kuala Lumpur 56000, Malaysia
| | - Sze Chun Chau
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Ka Chun Tsui
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Lee Wei Lim
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Kah Hui Wong
- Department of Anatomy, Faculty of Medicine, Universiti Malaya, Kuala Lumpur 50603, Malaysia
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| |
Collapse
|
7
|
Gartz M, Haberman M, Sutton J, Slick RA, Luttrell SM, Mack DL, Lawlor MW. ACTA1 H40Y mutant iPSC-derived skeletal myocytes display mitochondrial defects in an in vitro model of nemaline myopathy. Exp Cell Res 2023; 424:113507. [PMID: 36796746 PMCID: PMC9993434 DOI: 10.1016/j.yexcr.2023.113507] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 02/06/2023] [Accepted: 02/07/2023] [Indexed: 02/16/2023]
Abstract
Nemaline myopathies (NM) are a group of congenital myopathies that lead to muscle weakness and dysfunction. While 13 genes have been identified to cause NM, over 50% of these genetic defects are due to mutations in nebulin (NEB) and skeletal muscle actin (ACTA1), which are genes required for normal assembly and function of the thin filament. NM can be distinguished on muscle biopsies due to the presence of nemaline rods, which are thought to be aggregates of the dysfunctional protein. Mutations in ACTA1 have been associated with more severe clinical disease and muscle weakness. However, the cellular pathogenesis linking ACTA1 gene mutations to muscle weakness are unclear To evaluate cellular disease phenotypes, iPSC-derived skeletal myocytes (iSkM) harboring an ACTA1 H40Y point mutation were used to model NM in skeletal muscle. These were generated by Crispr-Cas9, and include one non-affected healthy control (C) and 2 NM iPSC clone lines, therefore representing isogenic controls. Fully differentiated iSkM were characterized to confirm myogenic status and subject to assays to evaluate nemaline rod formation, mitochondrial membrane potential, mitochondrial permeability transition pore (mPTP) formation, superoxide production, ATP/ADP/phosphate levels and lactate dehydrogenase release. C- and NM-iSkM demonstrated myogenic commitment as evidenced by mRNA expression of Pax3, Pax7, MyoD, Myf5 and Myogenin; and protein expression of Pax4, Pax7, MyoD and MF20. No nemaline rods were observed with immunofluorescent staining of NM-iSkM for ACTA1 or ACTN2, and these mRNA transcript and protein levels were comparable to C-iSkM. Mitochondrial function was altered in NM, as evidenced by decreased cellular ATP levels and altered mitochondrial membrane potential. Oxidative stress induction revealed the mitochondrial phenotype, as evidenced by collapsed mitochondrial membrane potential, early formation of the mPTP and increased superoxide production. Early mPTP formation was rescued with the addition of ATP to media. Together, these findings suggest that mitochondrial dysfunction and oxidative stress are disease phenotypes in the in vitro model of ACTA1 nemaline myopathy, and that modulation of ATP levels was sufficient to protect NM-iSkM mitochondria from stress-induced injury. Importantly, the nemaline rod phenotype was absent in our in vitro model of NM. We conclude that this in vitro model has the potential to recapitulate human NM disease phenotypes, and warrants further study.
Collapse
Affiliation(s)
- Melanie Gartz
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA.
| | - Margaret Haberman
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA; Diverge Translational Science Laboratory, Milwaukee, WI, USA
| | - Jessica Sutton
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA; Diverge Translational Science Laboratory, Milwaukee, WI, USA
| | - Rebecca A Slick
- Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Shawn M Luttrell
- Curi Bio Inc., 3000 Western Avenue, Seattle, WA, 98121, USA; Institute for Stem Cell and Regenerative Medicine, UW Medicine, Seattle, WA, USA
| | - David L Mack
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA; Institute for Stem Cell and Regenerative Medicine, UW Medicine, Seattle, WA, USA
| | - Michael W Lawlor
- Department of Cell Biology, Neurobiology & Anatomy, Medical College of Wisconsin, Milwaukee, WI, USA; Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, WI, USA; Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA; Diverge Translational Science Laboratory, Milwaukee, WI, USA
| |
Collapse
|
8
|
Di Leo V, Lawless C, Roussel MP, Gomes TB, Gorman GS, Russell OM, Tuppen HA, Duchesne E, Vincent AE. Resistance Exercise Training Rescues Mitochondrial Dysfunction in Skeletal Muscle of Patients with Myotonic Dystrophy Type 1. J Neuromuscul Dis 2023; 10:1111-1126. [PMID: 37638448 PMCID: PMC10657683 DOI: 10.3233/jnd-230099] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2023] [Indexed: 08/29/2023]
Abstract
BACKGROUND Myotonic dystrophy type 1 (DM1) is a dominant autosomal neuromuscular disorder caused by the inheritance of a CTG triplet repeat expansion in the Dystrophia Myotonica Protein Kinase (DMPK) gene. At present, no cure currently exists for DM1 disease. OBJECTIVE This study investigates the effects of 12-week resistance exercise training on mitochondrial oxidative phosphorylation in skeletal muscle in a cohort of DM1 patients (n = 11, men) in comparison to control muscle with normal oxidative phosphorylation. METHODS Immunofluorescence was used to assess protein levels of key respiratory chain subunits of complex I (CI) and complex IV (CIV), and markers of mitochondrial mass and cell membrane in individual myofibres sampled from muscle biopsies. Using control's skeletal muscle fibers population, we classified each patient's fibers as having normal, low or high levels of CI and CIV and compared the proportions of fibers before and after exercise training. The significance of changes observed between pre- and post-exercise within patients was estimated using a permutation test. RESULTS At baseline, DM1 patients present with significantly decreased mitochondrial mass, and isolated or combined CI and CIV deficiency. After resistance exercise training, in most patients a significant increase in mitochondrial mass was observed, and all patients showed a significant increase in CI and/or CIV protein levels. Moreover, improvements in mitochondrial mass were correlated with the one-repetition maximum strength evaluation. CONCLUSIONS Remarkably, 12-week resistance exercise training is sufficient to partially rescue mitochondrial dysfunction in DM1 patients, suggesting that the response to exercise is in part be due to changes in mitochondria.
Collapse
Affiliation(s)
- Valeria Di Leo
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, England
| | - Conor Lawless
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - Marie-Pier Roussel
- Department of Fundamental Sciences, Université du Québec à Chicoutimi, Quebec, Canada
| | - Tiago B. Gomes
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Royal Victoria Infirmary, Newcastle Upon Tyne, UK
| | - Gráinne S. Gorman
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, England
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Royal Victoria Infirmary, Newcastle Upon Tyne, UK
| | - Oliver M. Russell
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, England
| | - Helen A.L. Tuppen
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
| | - Elise Duchesne
- Department of Health Sciences, Université du Québec à Chicoutimi, Québec, Canada
- Neuromuscular Diseases Interdisciplinary Research Group (GRIMN), Saguenay-Lac-St-Jean Integrated University Health and Social Services Center, Saguenay, QC, Canada
| | - Amy E. Vincent
- Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK
- NIHR Newcastle Biomedical Research Centre, Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle upon Tyne, England
| |
Collapse
|
9
|
Shi X, Jiang X, Chen C, Zhang Y, Sun X. The interconnections between the microtubules and mitochondrial networks in cardiocerebrovascular diseases: Implications for therapy. Pharmacol Res 2022; 184:106452. [PMID: 36116706 DOI: 10.1016/j.phrs.2022.106452] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/13/2022] [Accepted: 09/13/2022] [Indexed: 10/14/2022]
Abstract
Microtubules, a highly dynamic cytoskeleton, participate in many cellular activities including mechanical support, organelles interactions, and intracellular trafficking. Microtubule organization can be regulated by modification of tubulin subunits, microtubule-associated proteins (MAPs) or agents modulating microtubule assembly. Increasing studies demonstrate that microtubule disorganization correlates with various cardiocerebrovascular diseases including heart failure and ischemic stroke. Microtubules also mediate intracellular transport as well as intercellular transfer of mitochondria, a power house in cells which produce ATP for various physiological activities such as cardiac mechanical function. It is known to all that both microtubules and mitochondria participate in the progression of cancer and Parkinson's disease. However, the interconnections between the microtubules and mitochondrial networks in cardiocerebrovascular diseases remain unclear. In this paper, we will focus on the roles of microtubules in cardiocerebrovascular diseases, and discuss the interplay of mitochondria and microtubules in disease development and treatment. Elucidation of these issues might provide significant diagnostic value as well as potential targets for cardiocerebrovascular diseases.
Collapse
Affiliation(s)
- Xingjuan Shi
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China.
| | - Xuan Jiang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Congwei Chen
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Yu Zhang
- School of Life Science and Technology, Key Laboratory of Developmental Genes and Human Disease, Southeast University, Nanjing, China
| | - Xiaoou Sun
- Institute of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou, China.
| |
Collapse
|
10
|
Yoshioka N, Kurose M, Yano M, Tran DM, Okuda S, Mori-Ochiai Y, Horie M, Nagai T, Nishino I, Shibata S, Takebayashi H. Isoform-specific mutation in Dystonin-b gene causes late-onset protein aggregate myopathy and cardiomyopathy. eLife 2022; 11:78419. [PMID: 35942699 PMCID: PMC9365387 DOI: 10.7554/elife.78419] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/18/2022] [Indexed: 12/03/2022] Open
Abstract
Dystonin (DST), which encodes cytoskeletal linker proteins, expresses three tissue-selective isoforms: neural DST-a, muscular DST-b, and epithelial DST-e. DST mutations cause different disorders, including hereditary sensory and autonomic neuropathy 6 (HSAN-VI) and epidermolysis bullosa simplex; however, etiology of the muscle phenotype in DST-related diseases has been unclear. Because DST-b contains all of the DST-a-encoding exons, known HSAN-VI mutations could affect both DST-a and DST-b isoforms. To investigate the specific function of DST-b in striated muscles, we generated a Dst-b-specific mutant mouse model harboring a nonsense mutation. Dst-b mutant mice exhibited late-onset protein aggregate myopathy and cardiomyopathy without neuropathy. We observed desmin aggregation, focal myofibrillar dissolution, and mitochondrial accumulation in striated muscles, which are common characteristics of myofibrillar myopathy. We also found nuclear inclusions containing p62, ubiquitin, and SUMO proteins with nuclear envelope invaginations as a unique pathological hallmark in Dst-b mutation-induced cardiomyopathy. RNA-sequencing analysis revealed changes in expression of genes responsible for cardiovascular functions. In silico analysis identified DST-b alleles with nonsense mutations in populations worldwide, suggesting that some unidentified hereditary myopathy and cardiomyopathy are caused by DST-b mutations. Here, we demonstrate that the Dst-b isoform is essential for long-term maintenance of striated muscles.
Collapse
Affiliation(s)
- Nozomu Yoshioka
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Transdisciplinary Research Programs, Niigata University, Niigata, Japan
| | - Masayuki Kurose
- Department of Physiology, School of Dentistry, Iwate Medical University, Iwate, Japan
| | - Masato Yano
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Dang Minh Tran
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Shujiro Okuda
- Medical AI Center, School of Medicine, Niigata University, Niigata, Japan
| | - Yukiko Mori-Ochiai
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masao Horie
- Department of Nursing, Niigata College of Nursing, Jōetsu, Japan
| | - Toshihiro Nagai
- Electron Microscope Laboratory, Keio University, Tokyo, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Shinsuke Shibata
- Electron Microscope Laboratory, Keio University, Tokyo, Japan.,Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Hirohide Takebayashi
- Division of Neurobiology and Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan.,Center for Coordination of Research Facilities, Niigata University, Niigata, Japan
| |
Collapse
|
11
|
Skeletal Muscle Mitochondria Dysfunction in Genetic Neuromuscular Disorders with Cardiac Phenotype. Int J Mol Sci 2021; 22:ijms22147349. [PMID: 34298968 PMCID: PMC8307986 DOI: 10.3390/ijms22147349] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 07/03/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial dysfunction is considered the major contributor to skeletal muscle wasting in different conditions. Genetically determined neuromuscular disorders occur as a result of mutations in the structural proteins of striated muscle cells and therefore are often combined with cardiac phenotype, which most often manifests as a cardiomyopathy. The specific roles played by mitochondria and mitochondrial energetic metabolism in skeletal muscle under muscle-wasting conditions in cardiomyopathies have not yet been investigated in detail, and this aspect of genetic muscle diseases remains poorly characterized. This review will highlight dysregulation of mitochondrial representation and bioenergetics in specific skeletal muscle disorders caused by mutations that disrupt the structural and functional integrity of muscle cells.
Collapse
|
12
|
Santhoshkumar R, Preethish-Kumar V, Polavarapu K, Reghunathan D, Chaudhari S, Satyamoorthy K, Vengalil S, Nashi S, Faruq M, Joshi A, Atchayaram N, Narayanappa G. A Novel L1 Linker Mutation in DES Resulted in Total Absence of Protein. J Mol Neurosci 2021; 71:2468-2473. [PMID: 34106405 DOI: 10.1007/s12031-021-01856-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Accepted: 05/06/2021] [Indexed: 11/29/2022]
Abstract
Desminopathies (MIM*601419) are clinically heterogeneous, manifesting with myopathy and/or cardiomyopathy and with intra-sarcoplasmic desmin-positive deposits. They have either an autosomal dominant (AD) or recessive (AR) pattern of inheritance. Desmin is a crucial intermediate filament protein regulating various cellular functions in muscle cells. Here, we report a 13-year-old girl, born of second-degree consanguineous parents, with normal developmental milestones, who presented with dilated cardiomyopathy, respiratory insufficiency and predominant distal upper limb weakness. A striking feature on muscle biopsy was the presence of a peripheral chain of nuclei in addition to myopathic features. Immunostaining showed complete lack of desmin expression, further confirmed by western blot analysis. Ultrastructurally, subsarcolemmal granular material, expanded Z-band aggregation, distortion of myofilaments, focal Z-band streaming, lobed and clustered myonuclei were observed. Next-generation sequencing revealed a novel homozygous nonsense mutation c.448C>T, p.R150X in the patient, while the parents were heterozygous carriers. Single mitochondrial DNA deletion and isolated complex IV deficiency were noted. Our findings add to the ever-expanding phenotype and molecular spectrum of desminopathies.
Collapse
Affiliation(s)
- Rashmi Santhoshkumar
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Veeramani Preethish-Kumar
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Kiran Polavarapu
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Dinesh Reghunathan
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Sima Chaudhari
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India
| | - Seena Vengalil
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Saraswati Nashi
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Muhammed Faruq
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110 007, India
| | - Aditi Joshi
- CSIR-Institute of Genomics and Integrative Biology, New Delhi, 110 007, India
| | - Nalini Atchayaram
- Department of Neurology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India
| | - Gayathri Narayanappa
- Department of Neuropathology, National Institute of Mental Health and Neuro Sciences, Bengaluru, Karnataka, 560 029, India.
| |
Collapse
|
13
|
Kalugina KK, Sukhareva KS, Churkinа AI, Kostareva AA. Autophagy as a Pathogenetic Link and
a Target for Therapy of Musculoskeletal System Diseases. J EVOL BIOCHEM PHYS+ 2021. [DOI: 10.1134/s0022093021030145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
14
|
Schänzer A, Schumann E, Zengeler D, Gulatz L, Maroli G, Ahting U, Sprengel A, Gräf S, Hahn A, Jux C, Acker T, Fürst DO, Rupp S, Schuld J, van der Ven PFM. The p.Ala2430Val mutation in filamin C causes a "hypertrophic myofibrillar cardiomyopathy". J Muscle Res Cell Motil 2021; 42:381-397. [PMID: 33710525 DOI: 10.1007/s10974-021-09601-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 02/26/2021] [Indexed: 10/21/2022]
Abstract
Hypertrophic cardiomyopathy (HCM) often leads to heart failure. Mutations in sarcomeric proteins are most frequently the cause of HCM but in many patients the gene defect is not known. Here we report on a young man who was diagnosed with HCM shortly after birth. Whole exome sequencing revealed a mutation in the FLNC gene (c.7289C > T; p.Ala2430Val) that was previously shown to cause aggregation of the mutant protein in transfected cells. Myocardial tissue from patients with this mutation has not been analyzed before and thus, the underlying etiology is not well understood. Myocardial tissue of our patient obtained during myectomy at the age of 23 years was analyzed in detail by histochemistry, immunofluorescence staining, electron microscopy and western blot analysis. Cardiac histology showed a pathology typical for myofibrillar myopathy with myofibril disarray and abnormal protein aggregates containing BAG3, desmin, HSPB5 and filamin C. Analysis of sarcomeric and intercalated disc proteins showed focally reduced expression of the gap junction protein connexin43 and Xin-positive sarcomeric lesions in the cardiomyocytes of our patient. In addition, autophagy pathways were altered with upregulation of LC3-II, WIPI1 and HSPB5, 6, 7 and 8. We conclude that the p.Ala2430Val mutation in FLNC most probably is associated with HCM characterized by abnormal intercalated discs, disarray of myofibrils and aggregates containing Z-disc proteins similar to myofibrillar myopathy, which supports the pathological effect of the mutation.
Collapse
Affiliation(s)
- Anne Schänzer
- Institute of Neuropathology, Justus Liebig University, Arndstr.16, 35392, Giessen, Germany.
| | - Elisabeth Schumann
- Institute of Neuropathology, Justus Liebig University, Arndstr.16, 35392, Giessen, Germany
| | - Diana Zengeler
- Center for Genomics and Transcriptomics (CeGat) GmbH, Tübingen, Germany
| | - Lisann Gulatz
- Institute of Neuropathology, Justus Liebig University, Arndstr.16, 35392, Giessen, Germany
| | - Giovanni Maroli
- Department of Cardiac Development and Remodeling, Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Uwe Ahting
- Institute of Human Genetics, Technical University of Munich (TUM), Munich, Germany
| | - Anke Sprengel
- Pediatric Heart Center, Justus Liebig University, Giessen, Germany
| | - Sabine Gräf
- Institute of Neuropathology, Justus Liebig University, Arndstr.16, 35392, Giessen, Germany
| | - Andreas Hahn
- Department of Child Neurology, Justus Liebig University, Giessen, Germany
| | - Christian Jux
- Pediatric Heart Center, Justus Liebig University, Giessen, Germany
| | - Till Acker
- Institute of Neuropathology, Justus Liebig University, Arndstr.16, 35392, Giessen, Germany
| | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Stefan Rupp
- Pediatric Heart Center, Justus Liebig University, Giessen, Germany
| | - Julia Schuld
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, Bonn, Germany
| |
Collapse
|
15
|
Qian FY, Guo YD, Zu J, Zhang JH, Zheng YM, Abdoulaye IA, Pan ZH, Xie CM, Gao HC, Zhang ZJ. A novel recessive mutation affecting DNAJB6a causes myofibrillar myopathy. Acta Neuropathol Commun 2021; 9:23. [PMID: 33557929 PMCID: PMC7869515 DOI: 10.1186/s40478-020-01046-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 10/02/2020] [Indexed: 11/10/2022] Open
Abstract
Mutations in the DNAJB6 gene have been identified as rare causes of myofibrillar myopathies. However, the underlying pathophysiologica mechanisms remain elusive. DNAJB6 has two known isoforms, including the nuclear isoform DNAJB6a and the cytoplasmic isoform DNAJB6b, which was thought to be the pathogenic isoform. Here, we report a novel recessive mutation c.695_699del (p. Val 232 Gly fs*7) in the DNAJB6 gene, associated with an apparently recessively inherited late onset distal myofibrillar myopathy in a Chinese family. Notably, the novel mutation localizes to exon 9 and uniquely encodes DNAJB6a. We further identified that this mutation decreases the mRNA and protein levels of DNAJB6a and results in an age-dependent recessive toxic effect on skeletal muscle in knock-in mice. Moreover, the mutant DNAJB6a showed a dose-dependent anti-aggregation effect on polyglutamine-containing proteins in vitro. Taking together, these findings reveal the pathogenic role of DNAJB6a insufficiency in myofibrillar myopathies and expand upon the molecular spectrum of DNAJB6 mutations.
Collapse
|
16
|
The Impact of Mitochondrial Deficiencies in Neuromuscular Diseases. Antioxidants (Basel) 2020; 9:antiox9100964. [PMID: 33050147 PMCID: PMC7600520 DOI: 10.3390/antiox9100964] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/23/2020] [Accepted: 09/24/2020] [Indexed: 02/06/2023] Open
Abstract
Neuromuscular diseases (NMDs) are a heterogeneous group of acquired or inherited rare disorders caused by injury or dysfunction of the anterior horn cells of the spinal cord (lower motor neurons), peripheral nerves, neuromuscular junctions, or skeletal muscles leading to muscle weakness and waste. Unfortunately, most of them entail serious or even fatal consequences. The prevalence rates among NMDs range between 1 and 10 per 100,000 population, but their rarity and diversity pose difficulties for healthcare and research. Some molecular hallmarks are being explored to elucidate the mechanisms triggering disease, to set the path for further advances. In fact, in the present review we outline the metabolic alterations of NMDs, mainly focusing on the role of mitochondria. The aim of the review is to discuss the mechanisms underlying energy production, oxidative stress generation, cell signaling, autophagy, and inflammation triggered or conditioned by the mitochondria. Briefly, increased levels of inflammation have been linked to reactive oxygen species (ROS) accumulation, which is key in mitochondrial genomic instability and mitochondrial respiratory chain (MRC) dysfunction. ROS burst, impaired autophagy, and increased inflammation are observed in many NMDs. Increasing knowledge of the etiology of NMDs will help to develop better diagnosis and treatments, eventually reducing the health and economic burden of NMDs for patients and healthcare systems.
Collapse
|
17
|
Decoding mitochondrial heterogeneity in single muscle fibres by imaging mass cytometry. Sci Rep 2020; 10:15336. [PMID: 32948797 PMCID: PMC7501294 DOI: 10.1038/s41598-020-70885-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 08/05/2020] [Indexed: 01/07/2023] Open
Abstract
The study of skeletal muscle continues to support the accurate diagnosis of mitochondrial disease and remains important in delineating molecular disease mechanisms. The heterogeneous expression of oxidative phosphorylation proteins and resulting respiratory deficiency are both characteristic findings in mitochondrial disease, hence the rigorous assessment of these at a single cell level is incredibly powerful. Currently, the number of proteins that can be assessed in individual fibres from a single section by immunohistochemistry is limited but imaging mass cytometry (IMC) enables the quantification of further, discrete proteins in individual cells. We have developed a novel workflow and bespoke analysis for applying IMC in skeletal muscle biopsies from patients with genetically-characterised mitochondrial disease, investigating the distribution of nine mitochondrial proteins in thousands of single muscle fibres. Using a semi-automated analysis pipeline, we demonstrate the accurate quantification of protein levels using IMC, providing an accurate measure of oxidative phosphorylation deficiency for complexes I-V at the single cell level. We demonstrate signatures of oxidative phosphorylation deficiency for common mtDNA variants and nuclear-encoded complex I variants and a compensatory upregulation of unaffected oxidative phosphorylation components. This technique can now be universally applied to evaluate a wide range of skeletal muscle disorders and protein targets.
Collapse
|
18
|
Alam S, Abdullah CS, Aishwarya R, Morshed M, Bhuiyan MS. Molecular Perspectives of Mitochondrial Adaptations and Their Role in Cardiac Proteostasis. Front Physiol 2020; 11:1054. [PMID: 32982788 PMCID: PMC7481364 DOI: 10.3389/fphys.2020.01054] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 07/31/2020] [Indexed: 12/17/2022] Open
Abstract
Mitochondria are the key to properly functioning energy generation in the metabolically demanding cardiomyocytes and thus essential to healthy heart contractility on a beat-to-beat basis. Mitochondria being the central organelle for cellular metabolism and signaling in the heart, its dysfunction leads to cardiovascular disease. The healthy mitochondrial functioning critical to maintaining cardiomyocyte viability and contractility is accomplished by adaptive changes in the dynamics, biogenesis, and degradation of the mitochondria to ensure cellular proteostasis. Recent compelling evidence suggests that the classical protein quality control system in cardiomyocytes is also under constant mitochondrial control, either directly or indirectly. Impairment of cytosolic protein quality control may affect the position of the mitochondria in relation to other organelles, as well as mitochondrial morphology and function, and could also activate mitochondrial proteostasis. Despite a growing interest in the mitochondrial quality control system, very little information is available about the molecular function of mitochondria in cardiac proteostasis. In this review, we bring together current understanding of the adaptations and role of the mitochondria in cardiac proteostasis and describe the adaptive/maladaptive changes observed in the mitochondrial network required to maintain proteomic integrity. We also highlight the key mitochondrial signaling pathways activated in response to proteotoxic stress as a cellular mechanism to protect the heart from proteotoxicity. A deeper understanding of the molecular mechanisms of mitochondrial adaptations and their role in cardiac proteostasis will help to develop future therapeutics to protect the heart from cardiovascular diseases.
Collapse
Affiliation(s)
- Shafiul Alam
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| | - Md Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States.,Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center, Shreveport, LA, United States
| |
Collapse
|
19
|
Multisystem Myotilinopathy, including Myopathy and Left Ventricular Noncompaction, due to the MYOT Variant c.179C>T. Case Rep Cardiol 2020; 2020:5128069. [PMID: 32509353 PMCID: PMC7244945 DOI: 10.1155/2020/5128069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Revised: 04/18/2020] [Accepted: 05/05/2020] [Indexed: 11/20/2022] Open
Abstract
Left ventricular hypertrabeculation/noncompaction is a myocardial abnormality of unknown etiology/pathogenesis, which is frequently associated with neuromuscular disorders or chromosomal defects. LVHT in association with a MYOT mutation has not been reported. The patient is a 72-year-old male with a history of strabismus in childhood, asymptomatic creatine-kinase elevation since age 42 years, slowly progressive lower limb weakness since age 60 years, slowly progressive dysarthria and dysphagia since age 62 years, and recurrent episodes of arthralgias and myalgias since age 71 years. He also had arterial hypertension, diverticulosis, hyperlipidemia, coronary heart disease, and a hiatal hernia with reflux esophagitis. Clinical exam revealed mild quadruparesis and proximal wasting of the legs. Whole exome sequencing revealed a known variant in the MYOT gene. Muscle biopsy, previously assessed as inclusion body myopathy, was compatible with the genotype after revision. Cardiologic work-up revealed a left anterior hemiblock, mild myocardial thickening, and noncompaction. This case shows that myotilinopathy may manifest as a multisystem disease, including noncompaction.
Collapse
|
20
|
Desminopathy: Novel Desmin Variants, a New Cardiac Phenotype, and Further Evidence for Secondary Mitochondrial Dysfunction. J Clin Med 2020; 9:jcm9040937. [PMID: 32235386 PMCID: PMC7231262 DOI: 10.3390/jcm9040937] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/20/2020] [Accepted: 03/24/2020] [Indexed: 12/13/2022] Open
Abstract
Background: The pleomorphic clinical presentation makes the diagnosis of desminopathy difficult. We aimed to describe the prevalence, phenotypic expression, and mitochondrial function of individuals with putative disease-causing desmin (DES) variants identified in patients with an unexplained etiology of cardiomyopathy. Methods: A total of 327 Czech patients underwent whole exome sequencing and detailed phenotyping in probands harboring DES variants. Results: Rare, conserved, and possibly pathogenic DES variants were identified in six (1.8%) probands. Two DES variants previously classified as variants of uncertain significance (p.(K43E), p.(S57L)), one novel DES variant (p.(A210D)), and two known pathogenic DES variants (p.(R406W), p.(R454W)) were associated with characteristic desmin-immunoreactive aggregates in myocardial and/or skeletal biopsy samples. The individual with the novel DES variant p.(Q364H) had a decreased myocardial expression of desmin with absent desmin aggregates in myocardial/skeletal muscle biopsy and presented with familial left ventricular non-compaction cardiomyopathy (LVNC), a relatively novel phenotype associated with desminopathy. An assessment of the mitochondrial function in four probands heterozygous for a disease-causing DES variant confirmed a decreased metabolic capacity of mitochondrial respiratory chain complexes in myocardial/skeletal muscle specimens, which was in case of myocardial succinate respiration more profound than in other cardiomyopathies. Conclusions: The presence of desminopathy should also be considered in individuals with LVNC, and in the differential diagnosis of mitochondrial diseases.
Collapse
|
21
|
Smolina N, Khudiakov A, Knyazeva A, Zlotina A, Sukhareva K, Kondratov K, Gogvadze V, Zhivotovsky B, Sejersen T, Kostareva A. Desmin mutations result in mitochondrial dysfunction regardless of their aggregation properties. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165745. [PMID: 32105824 DOI: 10.1016/j.bbadis.2020.165745] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 02/15/2020] [Accepted: 02/21/2020] [Indexed: 12/31/2022]
Abstract
Desmin, being a major intermediate filament of muscle cells, contributes to stabilization and positioning of mitochondria. Desmin mutations have been reported in conjunction with skeletal myopathies accompanied by mitochondrial dysfunction. Depending on the ability to promote intracellular aggregates formation, mutations can be considered aggregate-prone or non-aggregate-prone. The aim of the present study was to describe how expression of different desmin mutant isoforms effects mitochondria and contributes to the development of myocyte dysfunction. To achieve this goal, two non-aggregate-prone (Des S12F and Des A213V) and four aggregate-prone (Des L345P, Des A357P, Des L370P, Des D399Y) desmin mutations were expressed in skeletal muscle cells. We showed that all evaluated mutations affected the morphology of mitochondrial network, suppressed parameters of mitochondrial respiration, diminished mitochondrial membrane potential, increased ADP/ATP ratio, and enhanced mitochondrial DNA (mtDNA) release. mtDNA was partially secreted through exosomes as demonstrated by GW4869 treatment. Dysfunction of mitochondria was observed regardless the type of mutation: aggregate-prone or non-aggregate-prone. However, expression of aggregate-prone mutations resulted in more prominent phenotype. Thus, in this comparative study of six pathogenic desmin mutations that cause skeletal myopathy development, we confirmed a role of mitochondrial dysfunction and mtDNA release in the pathogenesis of desmin myopathies, regardless of the aggregation capacity of the mutated desmin.
Collapse
Affiliation(s)
- Natalia Smolina
- Almazov National Medical Research Centre, Saint Petersburg, Russia; Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden.
| | | | | | - Anna Zlotina
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Kseniya Sukhareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia; University of Verona, Verona, Italy
| | - Kirill Kondratov
- Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Vladimir Gogvadze
- Faculty of medicine, Lomonosov Moscow State University, Moscow, Russia; Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Boris Zhivotovsky
- Faculty of medicine, Lomonosov Moscow State University, Moscow, Russia; Institute of Environmental Medicine, Karolinska Institute, Stockholm, Sweden
| | - Thomas Sejersen
- Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russia; Department of Women's and Children's Health, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
22
|
Robertson R, Conte TC, Dicaire MJ, Rymar VV, Sadikot AF, Bryson-Richardson RJ, Lavoie JN, O'Ferrall E, Young JC, Brais B. BAG3 P215L/KO Mice as a Model of BAG3 P209L Myofibrillar Myopathy. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 190:554-562. [PMID: 31953038 DOI: 10.1016/j.ajpath.2019.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 11/22/2019] [Accepted: 11/26/2019] [Indexed: 10/25/2022]
Abstract
BCL-2-associated athanogene 3 (BAG3) is a co-chaperone to heat shock proteins important in degrading misfolded proteins through chaperone-assisted selective autophagy. The recurrent dominant BAG3-P209L mutation results in a severe childhood-onset myofibrillar myopathy (MFM) associated with progressive muscle weakness, cardiomyopathy, and respiratory failure. Because a homozygous knock-in (KI) strain for the mP215L mutation homologous to the human P209L mutation did not have a gross phenotype, compound heterozygote knockout (KO) and KI mP215L mice were generated to establish whether further reduction in BAG3 expression would lead to a phenotype. The KI/KO mice have a significant decrease in voluntary movement compared with wild-type and KI/KI mice in the open field starting at 7 months. The KI/KI and KI/KO mice both have significantly smaller muscle fiber cross-sectional area. However, only the KI/KO mice have clear skeletal muscle histologic changes in MFM. As in patient muscle, there are increased levels of BAG3-interacting proteins, such as p62, heat shock protein B8, and αB-crystallin. The KI/KO mP215L strain is the first murine model of BAG3 myopathy that resembles the human skeletal muscle pathologic features. The results support the hypothesis that the pathologic development of MFM requires a significant decrease in BAG3 protein level and not only a gain of function caused by the dominant missense mutation.
Collapse
Affiliation(s)
- Rebecca Robertson
- Neurogenetics of Motion Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Talita C Conte
- Neurogenetics of Motion Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada; Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Marie-Josée Dicaire
- Neurogenetics of Motion Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Vladimir V Rymar
- Cone Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Abbas F Sadikot
- Cone Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | | | - Josée N Lavoie
- Centre de Recherche sur le Cancer, l'Université Laval, Québec, Quebec, Canada; Oncology Axis, Centre de Recherche du Centre Hospitalier Universitaire (CHU), Québec-Université Laval, Québec, Quebec, Canada; Département de Biologie Moléculaire, Biochimie Médicale et Pathologie, l'Université Laval, Québec, Quebec, Canada
| | - Erin O'Ferrall
- Rare Neurological Diseases Group, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada
| | - Jason C Young
- Department of Biochemistry, McGill University, Montreal, Quebec, Canada
| | - Bernard Brais
- Neurogenetics of Motion Laboratory, Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
23
|
Mado K, Chekulayev V, Shevchuk I, Puurand M, Tepp K, Kaambre T. On the role of tubulin, plectin, desmin, and vimentin in the regulation of mitochondrial energy fluxes in muscle cells. Am J Physiol Cell Physiol 2019; 316:C657-C667. [PMID: 30811221 DOI: 10.1152/ajpcell.00303.2018] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Mitochondria perform a central role in life and death of the eukaryotic cell. They are major players in the generation of macroergic compounds and function as integrated signaling pathways, including the regulation of Ca2+ signals and apoptosis. A growing amount of evidence is demonstrating that mitochondria of muscle cells use cytoskeletal proteins (both microtubules and intermediate filaments) not only for their movement and proper cellular positioning, but also to maintain their biogenesis, morphology, function, and regulation of energy fluxes through the outer mitochondrial membrane (MOM). Here we consider the known literature data concerning the role of tubulin, plectin, desmin and vimentin in bioenergetic function of mitochondria in striated muscle cells, as well as in controlling the permeability of MOM for adenine nucleotides (ADNs). This is of great interest since dysfunctionality of these cytoskeletal proteins has been shown to result in severe myopathy associated with pronounced mitochondrial dysfunction. Further efforts are needed to uncover the pathways by which the cytoskeleton supports the functional capacity of mitochondria and transport of ADN(s) across the MOM (through voltage-dependent anion channel).
Collapse
Affiliation(s)
- Kati Mado
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Vladimir Chekulayev
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Igor Shevchuk
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Marju Puurand
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Kersti Tepp
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| | - Tuuli Kaambre
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics , Tallinn , Estonia
| |
Collapse
|
24
|
Tahrir FG, Langford D, Amini S, Mohseni Ahooyi T, Khalili K. Mitochondrial quality control in cardiac cells: Mechanisms and role in cardiac cell injury and disease. J Cell Physiol 2018; 234:8122-8133. [PMID: 30417391 DOI: 10.1002/jcp.27597] [Citation(s) in RCA: 171] [Impact Index Per Article: 24.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Accepted: 09/19/2018] [Indexed: 12/17/2022]
Abstract
Mitochondria play an important role in maintaining cardiac homeostasis by supplying the major energy required for cardiac excitation-contraction coupling as well as controlling the key intracellular survival and death pathways. Healthy mitochondria generate ATP molecules through an aerobic process known as oxidative phosphorylation (OXPHOS). Mitochondrial injury during myocardial infarction (MI) impairs OXPHOS and results in the excessive production of reactive oxygen species (ROS), bioenergetic insufficiency, and contributes to the development of cardiovascular diseases. Therefore, mitochondrial biogenesis along with proper mitochondrial quality control machinery, which removes unhealthy mitochondria is pivotal for mitochondrial homeostasis and cardiac health. Upon damage to the mitochondrial network, mitochondrial quality control components are recruited to segregate the unhealthy mitochondria and target aberrant mitochondrial proteins for degradation and elimination. Impairment of mitochondrial quality control and accumulation of abnormal mitochondria have been reported in the pathogenesis of various cardiac disorders and heart failure. Here, we provide an overview of the recent studies describing various mechanistic pathways underlying mitochondrial homeostasis with the main focus on cardiac cells. In addition, this review demonstrates the potential effects of mitochondrial quality control dysregulation in the development of cardiovascular disease.
Collapse
Affiliation(s)
- Farzaneh G Tahrir
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Dianne Langford
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Shohreh Amini
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Taha Mohseni Ahooyi
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| | - Kamel Khalili
- Department of Neuroscience, Center for Neurovirology, Lewis Katz School of Medicine at Temple University, Philadelphia, Pennsylvania
| |
Collapse
|
25
|
Isackson PJ, Wang J, Zia M, Spurgeon P, Levesque A, Bard J, James S, Nowak N, Lee TK, Vladutiu GD. RYR1 and CACNA1S genetic variants identified with statin-associated muscle symptoms. Pharmacogenomics 2018; 19:1235-1249. [PMID: 30325262 PMCID: PMC6563124 DOI: 10.2217/pgs-2018-0106] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 08/31/2018] [Indexed: 11/21/2022] Open
Abstract
AIM To examine the genetic differences between subjects with statin-associated muscle symptoms and statin-tolerant controls. MATERIALS & METHODS Next-generation sequencing was used to characterize the exomes of 76 subjects with severe statin-associated muscle symptoms and 50 statin-tolerant controls. RESULTS 12 probably pathogenic variants were found within the RYR1 and CACNA1S genes in 16% of cases with severe statin-induced myopathy representing a fourfold increase over variants found in statin-tolerant controls. Subjects with probably pathogenic RYR1 or CACNA1S variants had plasma CK 5X to more than 400X the upper limit of normal in addition to having muscle symptoms. CONCLUSIONS Genetic variants within the RYR1 and CACNA1S genes are likely to be a major contributor to the susceptibility to statin-associated muscle symptoms.
Collapse
Affiliation(s)
- Paul J Isackson
- Department of Pediatrics, State University of New York at Buffalo, NY 14203, USA
| | - Jianxin Wang
- Center for Computational Research, State University of New York at Buffalo, NY 14203, USA
| | - Mohammad Zia
- Center for Computational Research, State University of New York at Buffalo, NY 14203, USA
| | - Paul Spurgeon
- Center for Computational Research, State University of New York at Buffalo, NY 14203, USA
| | - Adrian Levesque
- Center for Computational Research, State University of New York at Buffalo, NY 14203, USA
| | - Jonathan Bard
- Center for Computational Research, State University of New York at Buffalo, NY 14203, USA
| | - Smitha James
- New York State Center of Excellence in Bioinformatics & Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Norma Nowak
- New York State Center of Excellence in Bioinformatics & Life Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
- Department of Biochemistry, Jacobs School of Medicine & Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY 14203, USA
| | - Tae Keun Lee
- Department of Pediatrics, State University of New York at Buffalo, NY 14203, USA
| | - Georgirene D Vladutiu
- Department of Pediatrics, State University of New York at Buffalo, NY 14203, USA
- Departments of Neurology & Pathology & Anatomical Sciences, University at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
26
|
Loss of CENP-F Results in Dilated Cardiomyopathy with Severe Disruption of Cardiac Myocyte Architecture. Sci Rep 2018; 8:7546. [PMID: 29765066 PMCID: PMC5953941 DOI: 10.1038/s41598-018-25774-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 04/27/2018] [Indexed: 12/14/2022] Open
Abstract
Centromere-binding protein F (CENP-F) is a very large and complex protein with many and varied binding partners including components of the microtubule network. Numerous CENP-F functions impacting diverse cellular behaviors have been identified. Importantly, emerging data have shown that CENP-F loss- or gain-of-function has critical effects on human development and disease. Still, it must be noted that data at the single cardiac myocyte level examining the impact of CENP-F loss-of-function on fundamental cellular behavior is missing. To address this gap in our knowledge, we analyzed basic cell structure and function in cardiac myocytes devoid of CENP-F. We found many diverse structural abnormalities including disruption of the microtubule network impacting critical characteristics of the cardiac myocyte. This is the first report linking microtubule network malfunction to cardiomyopathy. Importantly, we also present data demonstrating a direct link between a CENP-F single nucleotide polymorphism (snp) and human cardiac disease. In a proximate sense, these data examining CENP-F function explain the cellular basis underlying heart disease in this genetic model and, in a larger sense, they will hopefully provide a platform upon which the field can explore diverse cellular outcomes in wide-ranging areas of research on this critical protein.
Collapse
|
27
|
Murgia M, Toniolo L, Nagaraj N, Ciciliot S, Vindigni V, Schiaffino S, Reggiani C, Mann M. Single Muscle Fiber Proteomics Reveals Fiber-Type-Specific Features of Human Muscle Aging. Cell Rep 2018; 19:2396-2409. [PMID: 28614723 DOI: 10.1016/j.celrep.2017.05.054] [Citation(s) in RCA: 204] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 04/10/2017] [Accepted: 05/17/2017] [Indexed: 12/20/2022] Open
Abstract
Skeletal muscle is a key tissue in human aging, which affects different muscle fiber types unequally. We developed a highly sensitive single muscle fiber proteomics workflow to study human aging and show that the senescence of slow and fast muscle fibers is characterized by diverging metabolic and protein quality control adaptations. Whereas mitochondrial content declines with aging in both fiber types, glycolysis and glycogen metabolism are upregulated in slow but downregulated in fast muscle fibers. Aging mitochondria decrease expression of the redox enzyme monoamine oxidase A. Slow fibers upregulate a subset of actin and myosin chaperones, whereas an opposite change happens in fast fibers. These changes in metabolism and sarcomere quality control may be related to the ability of slow, but not fast, muscle fibers to maintain their mass during aging. We conclude that single muscle fiber analysis by proteomics can elucidate pathophysiology in a sub-type-specific manner.
Collapse
Affiliation(s)
- Marta Murgia
- Max-Planck-Institute of Biochemistry, Martinsried 82152, Germany; Department of Biomedical Science, University of Padova, Padua 35121, Italy.
| | - Luana Toniolo
- Department of Biomedical Science, University of Padova, Padua 35121, Italy
| | | | - Stefano Ciciliot
- Venetian Institute of Molecular Medicine, Padua 35129, Italy; Department of Medicine, University of Padua, Padua 35128, Italy
| | - Vincenzo Vindigni
- Department of Neurosciences, University of Padova, Padua 35128, Italy
| | | | - Carlo Reggiani
- Department of Biomedical Science, University of Padova, Padua 35121, Italy
| | - Matthias Mann
- Max-Planck-Institute of Biochemistry, Martinsried 82152, Germany.
| |
Collapse
|
28
|
Schänzer A, Rupp S, Gräf S, Zengeler D, Jux C, Akintürk H, Gulatz L, Mazhari N, Acker T, Van Coster R, Garvalov BK, Hahn A. Dysregulated autophagy in restrictive cardiomyopathy due to Pro209Leu mutation in BAG3. Mol Genet Metab 2018; 123:388-399. [PMID: 29338979 DOI: 10.1016/j.ymgme.2018.01.001] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 01/02/2018] [Accepted: 01/02/2018] [Indexed: 12/13/2022]
Abstract
Myofibrillary myopathies (MFM) are hereditary myopathies histologically characterized by degeneration of myofibrils and aggregation of proteins in striated muscle. Cardiomyopathy is common in MFM but the pathophysiological mechanisms are not well understood. The BAG3-Pro209Leu mutation is associated with early onset MFM and severe restrictive cardiomyopathy (RCM), often necessitating heart transplantation during childhood. We report on a young male patient with a BAG3-Pro209Leu mutation who underwent heart transplantation at eight years of age. Detailed morphological analyses of the explanted heart tissue showed intracytoplasmic inclusions, aggregation of BAG3 and desmin, disintegration of myofibers and Z-disk alterations. The presence of undegraded autophagosomes, seen by electron microscopy, as well as increased levels of p62, LC3-I and WIPI1, detected by immunohistochemistry and western blot analyses, indicated a dysregulation of autophagy. Parkin and PINK1, proteins involved in mitophagy, were slightly increased whereas mitochondrial OXPHOS activities were not altered. These findings indicate that altered autophagy plays a role in the pathogenesis and rapid progression of RCM in MFM caused by the BAG3-Pro209Leu mutation, which could have implications for future therapeutic strategies.
Collapse
Affiliation(s)
- A Schänzer
- Institute of Neuropathology, Justus Liebig University Giessen, 35392 Giessen, Germany.
| | - S Rupp
- Pediatric Heart Center, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - S Gräf
- Institute of Neuropathology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - D Zengeler
- Center for Genomics and Transcriptomics (CeGat) GmbH, 72076 Tübingen, Germany
| | - C Jux
- Pediatric Heart Center, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - H Akintürk
- Pediatric Heart Center, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - L Gulatz
- Institute of Neuropathology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - N Mazhari
- Pediatric Heart Center, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - T Acker
- Institute of Neuropathology, Justus Liebig University Giessen, 35392 Giessen, Germany
| | - R Van Coster
- Division of Child Neurology, Department of Pediatrics, University Hospital Gent, 9000 Gent, Belgium
| | - B K Garvalov
- Institute of Neuropathology, Justus Liebig University Giessen, 35392 Giessen, Germany; Department of Microvascular Biology and Pathobiology, Centre for Biomedicine and Medical Technology Mannheim (CBTM), Medical Faculty Mannheim, University of Heidelberg, 68167 Mannheim, Germany
| | - A Hahn
- Department of Child Neurology, Justus Liebig University Giessen, 35392 Giessen, Germany
| |
Collapse
|
29
|
Ramos-Fransi A, Martínez-Piñeiro A, Almendrote M, Lucente G, Carrato C, Ballester-Lopez A, Lucia A, Pintos-Morell G, Nogales-Gadea G, Coll-Cantí J. Myotilinopathy unmasked by statin treatment: A case report. Muscle Nerve 2018; 57:E138-E140. [PMID: 29350769 DOI: 10.1002/mus.26078] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2017] [Revised: 01/15/2018] [Accepted: 01/16/2018] [Indexed: 11/05/2022]
Affiliation(s)
- Alba Ramos-Fransi
- Neuromuscular Diseases Unit, Department of Neurosciences, Hospital Germans Trias i Pujol, Badalona, Spain.,Grup de Recerca en Malalties Neuromusculars i Neuropediatriques, Department of Neurosciences, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Alicia Martínez-Piñeiro
- Neuromuscular Diseases Unit, Department of Neurosciences, Hospital Germans Trias i Pujol, Badalona, Spain.,Grup de Recerca en Malalties Neuromusculars i Neuropediatriques, Department of Neurosciences, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Míriam Almendrote
- Neuromuscular Diseases Unit, Department of Neurosciences, Hospital Germans Trias i Pujol, Badalona, Spain.,Grup de Recerca en Malalties Neuromusculars i Neuropediatriques, Department of Neurosciences, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Giuseppe Lucente
- Neuromuscular Diseases Unit, Department of Neurosciences, Hospital Germans Trias i Pujol, Badalona, Spain.,Grup de Recerca en Malalties Neuromusculars i Neuropediatriques, Department of Neurosciences, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain
| | - Cristina Carrato
- Servicio de Anatomía Patológica, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Alfonsina Ballester-Lopez
- Grup de Recerca en Malalties Neuromusculars i Neuropediatriques, Department of Neurosciences, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Alejandro Lucia
- Instituto de Investigación Hospital 12 de Octubre (i+12), Madrid, Spain.,Universidad Europea de Madrid, Madrid, Spain
| | - Guillem Pintos-Morell
- Grup de Recerca en Malalties Neuromusculars i Neuropediatriques, Department of Neurosciences, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.,Servicio de Pediatría, Hospital Universitari Germans Trias i Pujol, Badalona, Spain
| | - Gisela Nogales-Gadea
- Grup de Recerca en Malalties Neuromusculars i Neuropediatriques, Department of Neurosciences, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Jaume Coll-Cantí
- Neuromuscular Diseases Unit, Department of Neurosciences, Hospital Germans Trias i Pujol, Badalona, Spain.,Grup de Recerca en Malalties Neuromusculars i Neuropediatriques, Department of Neurosciences, Institut d'Investigació en Ciències de la Salut Germans Trias i Pujol, Universitat Autònoma de Barcelona, Badalona, Spain.,Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
30
|
Alston CL, Rocha MC, Lax NZ, Turnbull DM, Taylor RW. The genetics and pathology of mitochondrial disease. J Pathol 2016; 241:236-250. [PMID: 27659608 PMCID: PMC5215404 DOI: 10.1002/path.4809] [Citation(s) in RCA: 294] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2016] [Revised: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 12/30/2022]
Abstract
Mitochondria are double-membrane-bound organelles that are present in all nucleated eukaryotic cells and are responsible for the production of cellular energy in the form of ATP. Mitochondrial function is under dual genetic control - the 16.6-kb mitochondrial genome, with only 37 genes, and the nuclear genome, which encodes the remaining ∼1300 proteins of the mitoproteome. Mitochondrial dysfunction can arise because of defects in either mitochondrial DNA or nuclear mitochondrial genes, and can present in childhood or adulthood in association with vast clinical heterogeneity, with symptoms affecting a single organ or tissue, or multisystem involvement. There is no cure for mitochondrial disease for the vast majority of mitochondrial disease patients, and a genetic diagnosis is therefore crucial for genetic counselling and recurrence risk calculation, and can impact on the clinical management of affected patients. Next-generation sequencing strategies are proving pivotal in the discovery of new disease genes and the diagnosis of clinically affected patients; mutations in >250 genes have now been shown to cause mitochondrial disease, and the biochemical, histochemical, immunocytochemical and neuropathological characterization of these patients has led to improved diagnostic testing strategies and novel diagnostic techniques. This review focuses on the current genetic landscape associated with mitochondrial disease, before focusing on advances in studying associated mitochondrial pathology in two, clinically relevant organs - skeletal muscle and brain. © 2016 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Charlotte L Alston
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Mariana C Rocha
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Nichola Z Lax
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Doug M Turnbull
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Robert W Taylor
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, The Medical School, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|