1
|
Wu W, Lv X, Feng Y, Yang M, Yang G, Zhao D, Yan C, Lin P. Mitochondrial dysfunction is driven by imbalanced fission and fusion of mitochondria in myofibrillar myopathy type 5. Hum Mol Genet 2025:ddaf051. [PMID: 40244302 DOI: 10.1093/hmg/ddaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 03/16/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
Myofibrillar myopathy type 5 (MFM5) is a dominantly inherited myopathy caused by mutations in the FLNC gene. The underlying pathogenic mechanisms of MFM5 remain unclear, and there are currently no effective treatments available. This study hypothesizes that mitochondrial dysfunction plays a key role in the pathogenesis of MFM5, on the basis of the COX-negative fibres observed in MFM5 patients. To test this hypothesis, a zebrafish model was developed to explore the impact of filamin-C on mitochondrial dynamics. These results demonstrated that defects in filamin-C disrupt mitochondrial fission, leading to mitochondrial dysfunction and mitophagy. This hypothesis was further validated through the analysis of skeletal muscle samples from MFM5 patients. These findings suggest that mitochondrial dysfunction caused by imbalanced fission and fusion of mitochondria and mitophagy contributes to MFM5 pathology. Importantly, this study identified potential therapeutic targets for MFM5 treatment, opening avenues for future research aimed at developing targeted interventions.
Collapse
Affiliation(s)
- Wenjing Wu
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Wenhua Xi Road 107, Jinan, Shandong 250012, China
| | - Xiaoqing Lv
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Wenhua Xi Road 107, Jinan, Shandong 250012, China
| | - Yifei Feng
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Wenhua Xi Road 107, Jinan, Shandong 250012, China
| | - Mengqi Yang
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Wenhua Xi Road 107, Jinan, Shandong 250012, China
| | - Guiguan Yang
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Wenhua Xi Road 107, Jinan, Shandong 250012, China
| | - Dandan Zhao
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Wenhua Xi Road 107, Jinan, Shandong 250012, China
| | - Chuanzhu Yan
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Wenhua Xi Road 107, Jinan, Shandong 250012, China
- Department of Neurology, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao 266000, China
- Mitochondrial Medicine Laboratory, Qilu Hospital (Qingdao), Cheeloo College of Medicine, Shandong University, 758 Hefei Road, Qingdao, Shandong 266035, China
- Brain Science Research Institute, Shandong University, Wenhua Xi Road 44, Jinan, Shandong 250000, China
| | - Pengfei Lin
- Department of Neurology, Shandong Key Laboratory of Mitochondrial Medicine and Rare Diseases, Research Institute of Neuromuscular and Neurodegenerative Diseases, Qilu Hospital of Shandong University, Wenhua Xi Road 107, Jinan, Shandong 250012, China
| |
Collapse
|
2
|
Holtzhausen C, Heil L, Klingel K, Fox H, Gummert J, Gärtner A, Schmidt A, Krüger M, Kirfel G, van der Ven PFM, Milting H, Clemen CS, Schröder R, Fürst DO, Tiesmeier J. Sudden cardiac death, arrhythmogenic cardiomyopathy and intercalated disc pathology due to reduced filamin C protein levels: a matter of life and death. Hum Mol Genet 2025; 34:726-738. [PMID: 39895064 DOI: 10.1093/hmg/ddaf014] [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: 11/05/2024] [Revised: 01/14/2025] [Accepted: 01/22/2025] [Indexed: 02/04/2025] Open
Abstract
Mutations in the human FLNC gene encoding filamin C (FLNc) cause a broad spectrum of sporadic and familial cardiomyopathies and myopathies. We report on the genetic, clinical, morphological and biochemical findings in a German family harboring an FLNC variant that leads to severe cardiac disease comprising sudden cardiac death and arrhythmogenic cardiomyopathy. Genetic analysis identified a novel heterozygous FLNC variant in exon 16 (NM_001458.4:c.2495_2498delAGTA, het; p.K832TfsX45) in i) the index patient suffering from dilated cardiomyopathy necessitating heart transplantation, ii) a son, who died from sudden cardiac death, iii) a second son, who survived an episode of sudden cardiac arrest and iv) a third son affected by isolated skeletal muscle myopathy. FLNc protein levels were markedly reduced in cardiac tissue obtained from the index patient, implying that the p.K832TfsX45 FLNc variant most probably caused nonsense-mediated decay of the corresponding mRNA. Morphological analysis of the diseased cardiac tissue revealed extensive fibrotic remodeling, and marked degenerative changes of the contractile apparatus of cardiomyocytes and severe structural alterations of intercalated discs. Connexin-43 signal intensity at intercalated discs was diminished and FLNc labelling of myofibrils was attenuated or even absent. Proteome analyses demonstrated complex alterations of extracellular matrix and intercalated disc proteins. Our findings demonstrate that this novel, truncating FLNC mutation likely leads to haploinsufficiency, thereby causing a deleterious sequence of degenerative changes of cardiac tissue with extensive fibrotic remodeling and intercalated disc pathology as the structural basis for FLNC-related cardiomyopathy with life-threatening cardiac arrhythmias.
Collapse
MESH Headings
- Female
- Humans
- Male
- Arrhythmias, Cardiac/genetics
- Arrhythmias, Cardiac/pathology
- Arrhythmogenic Right Ventricular Dysplasia/genetics
- Arrhythmogenic Right Ventricular Dysplasia/pathology
- Cardiomyopathy, Dilated/genetics
- Cardiomyopathy, Dilated/pathology
- Connexin 43/metabolism
- Connexin 43/genetics
- Death, Sudden, Cardiac/etiology
- Death, Sudden, Cardiac/pathology
- Filamins/genetics
- Filamins/metabolism
- Mutation
- Myocardium/pathology
- Myocardium/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Pedigree
Collapse
Affiliation(s)
- Christian Holtzhausen
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Lorena Heil
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, University Hospital Tübingen, Liebermeisterstr. 8, 72076 Tübingen, Germany
| | - Henrik Fox
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany
| | - Jan Gummert
- Clinic for Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany
| | - Anna Gärtner
- Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University of Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany
| | - Andreas Schmidt
- Center for Molecular Medicine (CMMC), Medical Faculty, and Excellence Cluster "Cellular Stress Responses in Aging-Associated Diseases" (CECAD), University of Cologne, Joseph-Stelzmann-Str. 26, 50937 Cologne, Germany
| | - Marcus Krüger
- Center for Molecular Medicine (CMMC), Medical Faculty, and Excellence Cluster "Cellular Stress Responses in Aging-Associated Diseases" (CECAD), University of Cologne, Joseph-Stelzmann-Str. 26, 50937 Cologne, Germany
| | - Gregor Kirfel
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Peter F M van der Ven
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University of Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany
| | - Christoph S Clemen
- Institute of Aerospace Medicine, German Aerospace Center (DLR), Linder Höhe, 51147 Cologne, Germany
- Institute of Vegetative Physiology, Medical Faculty, University of Cologne, Robert-Koch-Str. 39, 50931 Cologne, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Schwabachanlage 6, 91054 Erlangen, Germany
| | - Dieter O Fürst
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Jens Tiesmeier
- Erich and Hanna Klessmann Institute for Cardiovascular Research and Development, Heart and Diabetes Center North Rhine-Westphalia, Ruhr University of Bochum, Georgstr. 11, 32545 Bad Oeynhausen, Germany
- Institute for Anesthesiology, Intensive Care- and Emergency Medicine, MLK-Hospital, Voedestr. 79, Luebbecke, Campus OWL, Ruhr-University Bochum, 32312 Lübbecke, Germany
| |
Collapse
|
3
|
Kokot T, Zimmermann JP, Schwäble AN, Reimann L, Herr AL, Höfflin N, Köhn M, Warscheid B. Protein phosphatase-1 regulates the binding of filamin C to FILIP1 in cultured skeletal muscle cells under mechanical stress. Sci Rep 2024; 14:27348. [PMID: 39521905 PMCID: PMC11550807 DOI: 10.1038/s41598-024-78953-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
The actin-binding protein filamin c (FLNc) is a key mediator in the response of skeletal muscle cells to mechanical stress. In addition to its function as a structural scaffold, FLNc acts as a signaling adaptor which is phosphorylated at S2234 in its mechanosensitive domain 20 (d20) through AKT. Here, we discovered a strong dephosphorylation of FLNc-pS2234 in cultured skeletal myotubes under acute mechanical stress, despite high AKT activity. We found that all three protein phosphatase 1 (PP1) isoforms are part of the FLNc d18-21 interactome. Enzymatic assays demonstrate that PP1 efficiently dephosphorylates FLNc-pS2234 and in vitro and in cells upon PP1 activation using specific modulators. FLNc-pS2234 dephosphorylation promotes the interaction with FILIP1, a mediator for filamin degradation. Altogether, we present a model in which dephosphorylation of FLNc d20 by the dominant action of PP1c prevails over AKT activity to promote the binding of the filamin degradation-inducing factor FILIP1 during acute mechanical stress.
Collapse
Affiliation(s)
- Thomas Kokot
- Integrative Signaling Research, Institute of Biology III, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Johannes P Zimmermann
- Biochemistry II, Theodor-Boveri-Institut, Biozentrum, Faculty of Chemistry and Pharmacy, University of Würzburg, Würzburg, Germany
| | - Anja N Schwäble
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany
- Current address: Celonic AG, Basel, Switzerland
| | - Lena Reimann
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany
- Current address: Celonic AG, Basel, Switzerland
| | - Anna L Herr
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany
- Current address: Sartorius CellGenix GmbH, Freiburg, Germany
| | - Nico Höfflin
- Integrative Signaling Research, Institute of Biology III, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Maja Köhn
- Integrative Signaling Research, Institute of Biology III, University of Freiburg, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Bettina Warscheid
- Biochemistry II, Theodor-Boveri-Institut, Biozentrum, Faculty of Chemistry and Pharmacy, University of Würzburg, Würzburg, Germany.
- Biochemistry - Functional Proteomics, Institute of Biology II, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
4
|
Goliusova DV, Sharikova MY, Lavrenteva KA, Lebedeva OS, Muranova LK, Gusev NB, Bogomazova AN, Lagarkova MA. Role of Filamin C in Muscle Cells. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1546-1557. [PMID: 39418514 DOI: 10.1134/s0006297924090025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 07/02/2024] [Accepted: 08/14/2024] [Indexed: 10/19/2024]
Abstract
Filamin C (FLNC) is a member of a high-molecular weight protein family, which bind actin filaments in the cytoskeleton of various cells. In human genome FLNC is encoded by the FLNC gene located on chromosome 7 and is expressed predominantly in striated skeletal and cardiac muscle cells. Filamin C is involved in organization and stabilization of thin actin filaments three-dimensional network in sarcomeres, and is supposed to play a role of mechanosensor transferring mechanical signals to different protein targets. Under mechanical stress FLNC can undergo unfolding that increases the risk of its aggregation. FLNC molecules with an impaired native structure could be eliminated by the BAG3-mediated chaperone-assisted selective autophagy. Mutations in the FLNC gene could be accompanied by the changes in FLNC interaction with its protein partners and could lead to formation of aggregates, which overload the autophagy and proteasome protein degradation systems, thus facilitating development of various pathological processes. Molecular mechanisms of the FLNC-associated congenital disorders, called filaminopathies, remain poorly understood. This review is devoted to analysis of the structure and mechanisms of filamin C function in muscle and heart cells in normal state and in the FLNC-associated pathologies. The presented data summarize the results of research at the molecular, cellular, and tissue levels and allow us to outline promising ways for further investigation of pathogenetic mechanisms in filaminopathies.
Collapse
Affiliation(s)
- Daria V Goliusova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia.
- Koltzov Institute of Developmental Biology, Russian Academy of Sciences, Moscow, 119334, Russia
| | - Margarita Y Sharikova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Kristina A Lavrenteva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Olga S Lebedeva
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Lidia K Muranova
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Nikolai B Gusev
- Faculty of Biology, Lomonosov Moscow State University, Moscow, 119991, Russia
| | - Alexandra N Bogomazova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| | - Maria A Lagarkova
- Lopukhin Federal Research and Clinical Center of Physical-Chemical Medicine, Federal Medical Biological Agency, Moscow, 119435, Russia
| |
Collapse
|
5
|
Ohiri JC, Dellefave‐Castillo L, Tomar G, Wilsbacher L, Choudhury L, Barefield DY, Fullenkamp D, Gacita AM, Monroe TO, Pesce L, Blancard M, Vaught L, George AL, Demonbreun AR, Puckelwartz MJ, McNally EM. Reduction of Filamin C Results in Altered Proteostasis, Cardiomyopathy, and Arrhythmias. J Am Heart Assoc 2024; 13:e030467. [PMID: 38761081 PMCID: PMC11179814 DOI: 10.1161/jaha.123.030467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 04/17/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Many cardiomyopathy-associated FLNC pathogenic variants are heterozygous truncations, and FLNC pathogenic variants are associated with arrhythmias. Arrhythmia triggers in filaminopathy are incompletely understood. METHODS AND RESULTS We describe an individual with biallelic FLNC pathogenic variants, p.Arg650X and c.970-4A>G, with peripartum cardiomyopathy and ventricular arrhythmias. We also describe clinical findings in probands with FLNC variants including Val2715fs87X, Glu2458Serfs71X, Phe106Leu, and c.970-4A>G with hypertrophic and dilated cardiomyopathy, atrial fibrillation, and ventricular tachycardia. Induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) were generated. The FLNC truncation, Arg650X/c.970-4A>G, showed a marked reduction in filamin C protein consistent with biallelic loss of function mutations. To assess loss of filamin C, gene editing of a healthy control iPSC line was used to generate a homozygous FLNC disruption in the actin binding domain. Because filamin C has been linked to protein quality control, we assessed the necessity of filamin C in iPSC-CMs for response to the proteasome inhibitor bortezomib. After exposure to low-dose bortezomib, FLNC-null iPSC-CMs showed an increase in the chaperone proteins BAG3, HSP70 (heat shock protein 70), and HSPB8 (small heat shock protein B8) and in the autophagy marker LC3I/II. FLNC null iPSC-CMs had prolonged electric field potential, which was further prolonged in the presence of low-dose bortezomib. FLNC null engineered heart tissues had impaired function after low-dose bortezomib. CONCLUSIONS FLNC pathogenic variants associate with a predisposition to arrhythmias, which can be modeled in iPSC-CMs. Reduction of filamin C prolonged field potential, a surrogate for action potential, and with bortezomib-induced proteasome inhibition, reduced filamin C led to greater arrhythmia potential and impaired function.
Collapse
Affiliation(s)
- Joyce C. Ohiri
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | | | - Garima Tomar
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Lisa Wilsbacher
- Feinberg Cardiovascular and Renal Research Institute, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Lubna Choudhury
- Bluhm Cardiovascular InstituteNorthwestern MedicineChicagoILUSA
| | - David Y. Barefield
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
- Cell and Molecular PhysiologyLoyola University Stritch School of MedicineMaywoodILUSA
| | - Dominic Fullenkamp
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Anthony M. Gacita
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Tanner O. Monroe
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Lorenzo Pesce
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Malorie Blancard
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Lauren Vaught
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Alfred L. George
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Alexis R. Demonbreun
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Megan J. Puckelwartz
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
- Department of Pharmacology, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Feinberg School of MedicineNorthwestern UniversityChicagoILUSA
| |
Collapse
|
6
|
Onnée M, Bénézit A, Bastu S, Nadaj-Pakleza A, Lannes B, Ader F, Thèze C, Cintas P, Cances C, Carlier RY, Metay C, Cossée M, Malfatti E. The FLNC Ala1186Val Variant Linked to Cytoplasmic Body Myopathy and Cardiomyopathy Causes Protein Instability. Biomedicines 2024; 12:322. [PMID: 38397924 PMCID: PMC10887408 DOI: 10.3390/biomedicines12020322] [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: 12/29/2023] [Revised: 01/18/2024] [Accepted: 01/22/2024] [Indexed: 02/25/2024] Open
Abstract
Filamin C-related disorders include myopathies and cardiomyopathies linked to variants in the FLNC gene. Filamin C belongs to a family of actin-binding proteins involved in sarcomere stability. This study investigates the pathogenic impact of the FLNC c.3557C > T (p.Ala1186Val) pathogenic variant associated with an early-onset cytoplasmic body myopathy and cardiomyopathy in three unrelated patients. We performed clinical imaging and myopathologic and genetic characterization of three patients with an early-onset myopathy and cardiomyopathy. Bioinformatics analysis, variant interpretation, and protein structure analysis were performed to validate and assess the effects of the filamin C variant. All patients presented with a homogeneous clinical phenotype marked by a severe contractural myopathy, leading to loss of gait. There was prominent respiratory involvement and restrictive or hypertrophic cardiomyopathies. The Ala1186Val variant is located in the interstrand loop involved in intradomain stabilization and/or interdomain interactions with neighbor Ig-like domains. 3D modeling highlights local structural changes involving nearby residues and probably impacts the protein stability, causing protein aggregation in the form of cytoplasmic bodies. Myopathologic studies have disclosed the prominent aggregation and upregulation of the aggrephagy-associated proteins LC3B and p62. As a whole, the Ala1186Val variant in the FLNC gene provokes a severe myopathy with contractures, respiratory involvement, and cardiomyopathy due to protein aggregation in patients' muscles.
Collapse
Affiliation(s)
- Marion Onnée
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale U955, 94010 Créteil, France; (M.O.); (S.B.)
| | - Audrey Bénézit
- Neurologie et Réanimation Pédiatrique, Assistance Publique–Hôpitaux de Paris, Université Paris Saclay, Département Médico-Universitaire Santé de l’Enfant et de l’Adolescent, Hôpital Raymond Poincaré, 92380 Garches, France;
| | - Sultan Bastu
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale U955, 94010 Créteil, France; (M.O.); (S.B.)
| | - Aleksandra Nadaj-Pakleza
- Centre de Référence des Maladies Neuromusculaires Nord Est Ile-de-France, Service de Neurologie, Hôpitaux Universitaires de Strasbourg, 67200 Strasbourg, France;
- European Reference Network, EURO-NMD, Neuromuscular Centre at Hautepierre Hospital, Hôpitaux Universitaires de Strasbourg, 67200 Strasbourg, France
| | - Béatrice Lannes
- Département de Pathologie, Hôpitaux Universitaires de Strasbourg, 67091 Strasbourg, France;
| | - Flavie Ader
- Assistance Publique–Hôpitaux de Paris, Sorbonne Université, Département Médico-Universitaire BioGem, Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, 75013 Paris, France;
- Institut National de la Santé et de la Recherche Médicale UMRS1166, Université Paris Cité, 75006 Paris, France
| | - Corinne Thèze
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, Université de Montpellier, 34095 Montpellier, France;
| | - Pascal Cintas
- Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbes), Département de Neurologie, Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (P.C.); (M.C.)
| | - Claude Cances
- Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbes), Unité de Neurologie Pédiatrique, Hôpital des Enfants, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Robert-Yves Carlier
- Assistance Publique–Hôpitaux de Paris, Groupe Hospitalier Universitaire Paris Saclay, Département Médico-Universitaire Smart Imaging, Service d’Imagerie Médicale, Institut National de la Santé et de la Recherche Médicale UMR1179, Hôpital Raymond Poincaré, 92380 Garches, France;
| | - Corinne Metay
- Unité Fonctionnelle de Cardiogénétique et Myogénétique Moléculaire et Cellulaire, Centre de Génétique Moléculaire et Chromosomique, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Institut de Myologie, Groupe Hospitalier La Pitié-Salpêtrière, 75013 Paris, France;
| | - Mireille Cossée
- Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbes), Département de Neurologie, Hôpital Pierre-Paul Riquet, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (P.C.); (M.C.)
- PhyMedExp, Université de Montpellier, Institut National de la Santé et de la Recherche Médicale, Centre National de la Recherche Scientifique, 34295 Montpellier, France
| | - Edoardo Malfatti
- Institut Mondor de Recherche Biomédicale, Université Paris Est Créteil, Institut National de la Santé et de la Recherche Médicale U955, 94010 Créteil, France; (M.O.); (S.B.)
- Assistance Publique–Hôpitaux de Paris, Centre de Référence de Pathologie Neuromusculaire Nord-Est-Ile-de-France, Hôpital Henri Mondor, 94000 Créteil, France
| |
Collapse
|
7
|
Stonadge A, Genzor AV, Russell A, Hamed MF, Romero N, Evans G, Pownall ME, Bekker-Jensen S, Blanco G. Myofibrillar myopathy hallmarks associated with ZAK deficiency. Hum Mol Genet 2023; 32:2751-2770. [PMID: 37427997 PMCID: PMC10789240 DOI: 10.1093/hmg/ddad113] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 07/04/2023] [Accepted: 07/06/2023] [Indexed: 07/11/2023] Open
Abstract
The ZAK gene encodes two functionally distinct kinases, ZAKα and ZAKβ. Homozygous loss of function mutations affecting both isoforms causes a congenital muscle disease. ZAKβ is the only isoform expressed in skeletal muscle and is activated by muscle contraction and cellular compression. The ZAKβ substrates in skeletal muscle or the mechanism whereby ZAKβ senses mechanical stress remains to be determined. To gain insights into the pathogenic mechanism, we exploited ZAK-deficient cell lines, zebrafish, mice and a human biopsy. ZAK-deficient mice and zebrafish show a mild phenotype. In mice, comparative histopathology data from regeneration, overloading, ageing and sex conditions indicate that while age and activity are drivers of the pathology, ZAKβ appears to have a marginal role in myoblast fusion in vitro or muscle regeneration in vivo. The presence of SYNPO2, BAG3 and Filamin C (FLNC) in a phosphoproteomics assay and extended analyses suggested a role for ZAKβ in the turnover of FLNC. Immunofluorescence analysis of muscle sections from mice and a human biopsy showed evidence of FLNC and BAG3 accumulations as well as other myofibrillar myopathy markers. Moreover, endogenous overloading of skeletal muscle exacerbated the presence of fibres with FLNC accumulations in mice, indicating that ZAKβ signalling is necessary for an adaptive turnover of FLNC that allows for the normal physiological response to sustained mechanical stress. We suggest that accumulation of mislocalized FLNC and BAG3 in highly immunoreactive fibres contributes to the pathogenic mechanism of ZAK deficiency.
Collapse
Affiliation(s)
- Amy Stonadge
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5DD, UK
| | - Aitana V Genzor
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Alex Russell
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5DD, UK
| | - Mohamed F Hamed
- Department of Pathology, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Norma Romero
- Unité de Morphologie Neuromusculaire Institut de Myologie - Inserm Sorbonne Université - GHU Pitié-Salpêtrière 47- 83, boulevard de l’Hôpital F-75 651 Paris, Cedex 13, France
| | - Gareth Evans
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5DD, UK
| | - Mary Elizabeth Pownall
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5DD, UK
| | - Simon Bekker-Jensen
- Center for Healthy Aging, Department of Cellular and Molecular Medicine, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Gonzalo Blanco
- York Biomedical Research Institute, Department of Biology, University of York, York, YO10 5DD, UK
| |
Collapse
|
8
|
Sellung D, Heil L, Daya N, Jacobsen F, Mertens-Rill J, Zhuge H, Döring K, Piran M, Milting H, Unger A, Linke WA, Kley R, Preusse C, Roos A, Fürst DO, Ven PFMVD, Vorgerd M. Novel Filamin C Myofibrillar Myopathy Variants Cause Different Pathomechanisms and Alterations in Protein Quality Systems. Cells 2023; 12:cells12091321. [PMID: 37174721 PMCID: PMC10177260 DOI: 10.3390/cells12091321] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 04/28/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
Myofibrillar myopathies (MFM) are a group of chronic muscle diseases pathophysiologically characterized by accumulation of protein aggregates and structural failure of muscle fibers. A subtype of MFM is caused by heterozygous mutations in the filamin C (FLNC) gene, exhibiting progressive muscle weakness, muscle structural alterations and intracellular protein accumulations. Here, we characterize in depth the pathogenicity of two novel truncating FLNc variants (p.Q1662X and p.Y2704X) and assess their distinct effect on FLNc stability and distribution as well as their impact on protein quality system (PQS) pathways. Both variants cause a slowly progressive myopathy with disease onset in adulthood, chronic myopathic alterations in muscle biopsy including the presence of intracellular protein aggregates. Our analyses revealed that p.Q1662X results in FLNc haploinsufficiency and p.Y2704X in a dominant-negative FLNc accumulation. Moreover, both protein-truncating variants cause different PQS alterations: p.Q1662X leads to an increase in expression of several genes involved in the ubiquitin-proteasome system (UPS) and the chaperone-assisted selective autophagy (CASA) system, whereas p.Y2704X results in increased abundance of proteins involved in UPS activation and autophagic buildup. We conclude that truncating FLNC variants might have different pathogenetic consequences and impair PQS function by diverse mechanisms and to varying extents. Further studies on a larger number of patients are necessary to confirm our observations.
Collapse
Affiliation(s)
- Dominik Sellung
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Lorena Heil
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Nassam Daya
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Frank Jacobsen
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Janine Mertens-Rill
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Heidi Zhuge
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Kristina Döring
- Department of Human Genetics, Ruhr-University Bochum, 44801 Bochum, Germany
| | - Misagh Piran
- Erich and Hanna Klessmann Institute, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Centre NRW, University Hospital of the Ruhr-University Bochum, 32545 Bad Oeynhausen, Germany
| | - Andreas Unger
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Wolfgang A Linke
- Institute of Physiology II, University of Münster, 48149 Münster, Germany
| | - Rudi Kley
- Department of Neurology and Clinical Neurophysiology, St. Marien-Hospital Borken, 46325 Borken, Germany
| | - Corinna Preusse
- Department of Neuropathology, Charité-Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Andreas Roos
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121 Bonn, Germany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, 44789 Bochum, Germany
| |
Collapse
|
9
|
Noureddine M, Gehmlich K. Structural and signaling proteins in the Z-disk and their role in cardiomyopathies. Front Physiol 2023; 14:1143858. [PMID: 36935760 PMCID: PMC10017460 DOI: 10.3389/fphys.2023.1143858] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/21/2023] [Indexed: 03/06/2023] Open
Abstract
The sarcomere is the smallest functional unit of muscle contraction. It is delineated by a protein-rich structure known as the Z-disk, alternating with M-bands. The Z-disk anchors the actin-rich thin filaments and plays a crucial role in maintaining the mechanical stability of the cardiac muscle. A multitude of proteins interact with each other at the Z-disk and they regulate the mechanical properties of the thin filaments. Over the past 2 decades, the role of the Z-disk in cardiac muscle contraction has been assessed widely, however, the impact of genetic variants in Z-disk proteins has still not been fully elucidated. This review discusses the various Z-disk proteins (alpha-actinin, filamin C, titin, muscle LIM protein, telethonin, myopalladin, nebulette, and nexilin) and Z-disk-associated proteins (desmin, and obscurin) and their role in cardiac structural stability and intracellular signaling. This review further explores how genetic variants of Z-disk proteins are linked to inherited cardiac conditions termed cardiomyopathies.
Collapse
Affiliation(s)
- Maya Noureddine
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
| | - Katja Gehmlich
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, United Kingdom
- Cardiovascular Medicine, Radcliffe Department of Medicine and British Heart Foundation Centre of Research Excellence Oxford, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
10
|
Abstract
Inherited myopathies are a group of disease, which, although distinct from a genetic and prognostic point of view, can lead to non-specific clinical pictures due to phenotypic overlap. Acquired immuno-mediated myopathies may also pose the problem of clinically accurate etiological orientation. The assessment of fatty infiltration and pathological increase in water volume of the muscle contingent on whole-body muscle MRI is becoming increasingly important in aiding the initial diagnosis of inherited and acquired myopathies. MRI helps orientating the clinical diagnostic hypotheses thanks to the patterns of muscle involved (more or less specific according to the entities), which led to the development of decision-making algorithms proposed in the literature. The aim of this article is to specify the proper MRI protocol for the evaluation of myopathies and the basis of the interpretation and to provide a summary of the most frequently inherited and acquired myopathies described in the literature.
Collapse
|
11
|
Wei XJ, Sun H, Miao J, Qiu RQ, Jiang ZZ, Ma ZW, Sun W, Yu XF. Clinical-pathological features and muscle imaging findings in 36 Chinese patients with rimmed vacuolar myopathies: case series study and review of literature. Front Neurol 2023; 14:1152738. [PMID: 37188302 PMCID: PMC10175607 DOI: 10.3389/fneur.2023.1152738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/03/2023] [Indexed: 05/17/2023] Open
Abstract
Introduction Rimmed vacuolar myopathies (RVMs) are a group of genetically heterogeneous diseases that share histopathological characteristics on muscle biopsy, including the aberrant accumulation of autophagic vacuoles. However, the presence of non-coding sequences and structural mutations, some of which remain undetectable, confound the identification of pathogenic mutations responsible for RVMs. Therefore, we assessed the clinical profiles and muscle magnetic resonance imaging (MRI) changes in 36 Chinese patients with RVMs, emphasizing the role of muscle MRI in disease identification and differential diagnosis to propose a comprehensive literature-based imaging pattern to facilitate improved diagnostic workup. Methods All patients presented with rimmed vacuoles with varying degrees of muscular dystrophic changes and underwent a comprehensive evaluation using clinical, morphological, muscle MRI and molecular genetic analysis. We assessed muscle changes in the Chinese RVMs and provided an overview of the RVMs, focusing on the patterns of muscle involvement on MRI. Results A total of 36 patients, including 24 with confirmed distal myopathy and 12 with limb-girdle phenotype, had autophagic vacuoles with RVMs. Hierarchical clustering of patients according to the predominant effect of the distal or proximal lower limbs revealed that most patients with RVMs could be distinguished. GNE myopathy was the most prevalent form of RVMs observed in this study. Moreover, MRI helped identify the causative genes in some diseases (e.g., desminopathy and hereditary myopathy with early respiratory failure) and confirmed the pathogenicity of a novel mutation (e.g., adult-onset proximal rimmed vacuolar titinopathy) detected using next-generation sequencing. Discussion Collectively, our findings expand our knowledge of the genetic spectrum of RVMs in China and suggest that muscle imaging should be an integral part of assisting genetic testing and avoiding misdiagnosis in the diagnostic workup of RVM.
Collapse
|
12
|
Velardo D, D'Angelo MG, Citterio A, Panzeri E, Napoli L, Cinnante C, Moggio M, Comi GP, Ronchi D, Bassi MT. Case Reports: Novel Missense Variants in the Filamin C Actin Binding Domain Cause Variable Phenotypes. Front Neurol 2022; 13:930039. [PMID: 35903116 PMCID: PMC9315448 DOI: 10.3389/fneur.2022.930039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 06/22/2022] [Indexed: 11/24/2022] Open
Abstract
Filamin C is a large dimeric actin-binding protein, most prevalent in skeletal and cardiac muscle Z-discs, where it participates in sarcomere mechanical stabilization and intracellular signaling, interacting with numerous binding partners. Dominant heterozygous mutations of Filamin C gene cause several forms of myopathy and structural or arrhythmogenic cardiomyopathy. In this report we describe clinical and molecular findings of two Italian patients, in whom we identified two novel missense variants located within the Filamin C actin binding domain. Muscle imaging, histological and ultrastructural findings are also reported. Our results underline the extreme inter- and intrafamilial variability of clinical manifestations, hence the need to extend the investigation also to asymptomatic relatives, and the relevance of a broad diagnostic approach involving muscle electron microscopy, skeletal muscle magnetic resonance imaging and next generation sequencing techniques.
Collapse
Affiliation(s)
- Daniele Velardo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy
- *Correspondence: Daniele Velardo
| | - Maria Grazia D'Angelo
- NeuroMuscular Unit, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) E. Medea, Bosisio Parini, Italy
| | - Andrea Citterio
- Laboratory of Molecular Biology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) E. Medea, Bosisio Parini, Italy
| | - Elena Panzeri
- Laboratory of Molecular Biology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) E. Medea, Bosisio Parini, Italy
| | - Laura Napoli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy
| | - Claudia Cinnante
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuroradiology Unit, Milan, Italy
- Department of Radiology, Istituto Auxologico Italiano, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS), Milan, Italy
| | - Maurizio Moggio
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy
| | - Giacomo Pietro Comi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Neuromuscular and Rare Diseases Unit, Department of Neuroscience, Milan, Italy
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Neuroscience Section, University of Milan, Milan, Italy
| | - Dario Ronchi
- Department of Pathophysiology and Transplantation, Dino Ferrari Center, Neuroscience Section, University of Milan, Milan, Italy
| | - Maria Teresa Bassi
- Laboratory of Molecular Biology, Scientific Institute for Research, Hospitalization and Healthcare (IRCCS) E. Medea, Bosisio Parini, Italy
| |
Collapse
|
13
|
Huang C, Zheng Y, Zhang W, Chen Z, Huang Z, Fang Y. Case Report: A Chinese Family of Hypertrophic Cardiomyopathy Caused by a Novel Splicing Mutation in the FLNC Gene. Front Genet 2022; 13:894791. [PMID: 35795207 PMCID: PMC9251305 DOI: 10.3389/fgene.2022.894791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Accepted: 05/10/2022] [Indexed: 11/13/2022] Open
Abstract
Hypertrophic cardiomyopathy (HCM) is a type of primary cardiomyopathy with genetic etiology, and it carries a high risk of diastolic dysfunction, heart failure, and malignant arrhythmias. We reported the first familial HCM in China, caused by a novel FLNC splicing mutation. We performed duo exome sequencing (ES) to examine the genome of the proband and his mother. For 10 days, a 15-year-old boy was presented to our hospital due to non–exercise-associated chest tightness and asthma. He was diagnosed with HCM [end-diastolic interventricular septal thickness was about 18 mm by transthoracic echocardiography (TTE)]. His mother and sister performed TTE to screen familial cardiomyopathy, which revealed hypertrophic cardiomyopathy only in the proband’s mother. In ES of the mother–son duo, we identified a novel heterozygous mutation of the FLNC gene (chr7:128492808, NM_001127487, c.5905+2T>C, rs1808874360) as the candidate cause of autosomal dominant HCM. Sanger sequencing confirmed this novel mutation in the proband and his mother but absent in the proband’s sister. The potential impact of the novel mutation was predicted by MutationTaster, dbscSNV_ADA_SCORE, dbscSNV_RF_SCORE, CADD_phred, PhyloP20way_mammalian, PhyloP100way_vertebrate, SiPhy_29way_logOdds, and GERP++_RS software. After the administration of furosemide, spironolactone, and metoprolol, the proband’s heart function was improved, and symptoms were alleviated. We presented the first familial HCM caused by a novel FLNC splicing mutation via exome sequencing in China. Therefore, it is necessary that familial screening for patients with HCM should be performed for the early detection of HCM intervention in malignant cardiac events in advance and block genes.
Collapse
Affiliation(s)
- Chunhui Huang
- Department of Cardiology, Liyang City Hospital of Traditional Chinese Medicine, Liyang, China
| | - Yonghong Zheng
- Department of Cardiology, Liyang City Hospital of Traditional Chinese Medicine, Liyang, China
| | - Wei Zhang
- Department of Cardiology, Liyang City Hospital of Traditional Chinese Medicine, Liyang, China
| | - Zhigang Chen
- Department of Cardiology, Liyang City Hospital of Traditional Chinese Medicine, Liyang, China
| | - Zhixin Huang
- Department of Ultrasound Medicine, Liyang City Hospital of Traditional Chinese Medicine, Liyang, China
| | - Yuan Fang
- Department of Geriatric Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Yuan Fang,
| |
Collapse
|
14
|
Rodina N, Khudiakov A, Perepelina K, Muravyev A, Boytsov A, Zlotina A, Sokolnikova P, Kostareva A. Generation of iPSC line (FAMRCi009-A) from patient with familial progressive cardiac conduction disorder carrying genetic variant FLNC p.Val2264Met. Stem Cell Res 2022; 59:102640. [PMID: 34971933 DOI: 10.1016/j.scr.2021.102640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 12/17/2021] [Accepted: 12/20/2021] [Indexed: 11/22/2022] Open
Abstract
Human iPSC cell line FAMRCi009-A was generated from a patient with restrictive cardiomyopathy and congenital myopathy carrying FLNC p.Val2264Met genetic variant. Patient-specific peripheral blood mononuclear cells were reprogrammed using non-integrative Sendai viruses. Generated iPSC lines showed normal karyotype, expressed pluripotency markers and exhibited trilineage differentiation potential in vitro. The reported iPSC lines could be used for a deeper study of filaminopathies.
Collapse
Affiliation(s)
- Nataliia Rodina
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | | | - Kseniya Perepelina
- Almazov National Medical Research Centre, Saint-Petersburg, Russia; Saint Petersburg State University, Saint-Petersburg, Russia.
| | - Aleksey Muravyev
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | - Aleksandr Boytsov
- Almazov National Medical Research Centre, Saint-Petersburg, Russia; ITMO University, Saint-Petersburg, Russia
| | - Anna Zlotina
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| | | | - Anna Kostareva
- Almazov National Medical Research Centre, Saint-Petersburg, Russia
| |
Collapse
|
15
|
Agarwal R, Paulo JA, Toepfer CN, Ewoldt JK, Sundaram S, Chopra A, Zhang Q, Gorham J, DePalma SR, Chen CS, Gygi SP, Seidman CE, Seidman JG. Filamin C Cardiomyopathy Variants Cause Protein and Lysosome Accumulation. Circ Res 2021; 129:751-766. [PMID: 34405687 PMCID: PMC9053646 DOI: 10.1161/circresaha.120.317076] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 08/17/2021] [Indexed: 01/02/2023]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Radhika Agarwal
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher N. Toepfer
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Radcliffe Department of Medicine, University of Oxford, OX3 9DU, UK
- Wellcome Centre for Human Genetics, University of Oxford, OX3 7BN, UK
| | - Jourdan K. Ewoldt
- Department of Biomedical Engineering, Boston University, Boston, MA 02115, USA
| | - Subramanian Sundaram
- Department of Biomedical Engineering, Boston University, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Anant Chopra
- Department of Biomedical Engineering, Boston University, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Qi Zhang
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Joshua Gorham
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Steven R. DePalma
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Christopher S. Chen
- Department of Biomedical Engineering, Boston University, Boston, MA 02115, USA
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Steven P. Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Christine E. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - J. G. Seidman
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
16
|
Abstract
Cardiomyopathy affects approximately 1 in 500 adults and is the leading cause of death. Familial cases are common, and mutations in many genes are involved in cardiomyopathy, especially those in genes encoding cytoskeletal, sarcomere, and nuclear envelope proteins. Filamin C is an actin-binding protein encoded by filamin C (FLNC) gene and participates in sarcomere stability maintenance. FLNC was first demonstrated to be a causal gene of myofibrillar myopathy; recently, it has been found that FLNC mutation plays a critical role in the pathogenesis of cardiomyopathy. In this review, we summarized the physiological roles of filamin C in cardiomyocytes and the genetic evidence for links between FLNC mutations and cardiomyopathies. Truncated FLNC is enriched in dilated cardiomyopathy and arrhythmogenic right ventricular cardiomyopathy. Non-truncated FLNC is enriched in hypertrophic cardiomyopathy and restrictive cardiomyopathy. Two major pathomechanisms in FLNC-related cardiomyopathy have been described: protein aggregation resulting from non-truncating mutations and haploinsufficiency triggered by filamin C truncation. Therefore, it is important to understand the cellular biology and molecular regulation of FLNC to design new therapies to treat patients with FLNC-related cardiomyopathy.
Collapse
|
17
|
Eggers B, Schork K, Turewicz M, Barkovits K, Eisenacher M, Schröder R, Clemen CS, Marcus K. Advanced Fiber Type-Specific Protein Profiles Derived from Adult Murine Skeletal Muscle. Proteomes 2021; 9:proteomes9020028. [PMID: 34201234 PMCID: PMC8293376 DOI: 10.3390/proteomes9020028] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/01/2021] [Accepted: 06/02/2021] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle is a heterogeneous tissue consisting of blood vessels, connective tissue, and muscle fibers. The last are highly adaptive and can change their molecular composition depending on external and internal factors, such as exercise, age, and disease. Thus, examination of the skeletal muscles at the fiber type level is essential to detect potential alterations. Therefore, we established a protocol in which myosin heavy chain isoform immunolabeled muscle fibers were laser microdissected and separately investigated by mass spectrometry to develop advanced proteomic profiles of all murine skeletal muscle fiber types. All data are available via ProteomeXchange with the identifier PXD025359. Our in-depth mass spectrometric analysis revealed unique fiber type protein profiles, confirming fiber type-specific metabolic properties and revealing a more versatile function of type IIx fibers. Furthermore, we found that multiple myopathy-associated proteins were enriched in type I and IIa fibers. To further optimize the assignment of fiber types based on the protein profile, we developed a hypothesis-free machine-learning approach, identified a discriminative peptide panel, and confirmed our panel using a public data set.
Collapse
Affiliation(s)
- Britta Eggers
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany; (K.S.); (M.T.); (K.B.); (M.E.)
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, 44801 Bochum, Germany
- Correspondence: (B.E.); (K.M.)
| | - Karin Schork
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany; (K.S.); (M.T.); (K.B.); (M.E.)
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, 44801 Bochum, Germany
| | - Michael Turewicz
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany; (K.S.); (M.T.); (K.B.); (M.E.)
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, 44801 Bochum, Germany
| | - Katalin Barkovits
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany; (K.S.); (M.T.); (K.B.); (M.E.)
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, 44801 Bochum, Germany
| | - Martin Eisenacher
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany; (K.S.); (M.T.); (K.B.); (M.E.)
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, 44801 Bochum, Germany
| | - Rolf Schröder
- Institute of Neuropathology, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, 91054 Erlangen, Germany;
| | - Christoph S. Clemen
- German Aerospace Center, Institute of Aerospace Medicine, 51147 Cologne, Germany;
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Medical Faculty, University of Cologne, 50931 Cologne, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Medical Faculty, Ruhr-University Bochum, 44801 Bochum, Germany; (K.S.); (M.T.); (K.B.); (M.E.)
- Medical Proteome Analysis, Center for Protein Diagnostics (PRODI), Ruhr-University Bochum, 44801 Bochum, Germany
- Correspondence: (B.E.); (K.M.)
| |
Collapse
|
18
|
Liewluck T. A Window Into the Myofibrillar Myopathy Proteome. Neurol Genet 2021; 7:e587. [PMID: 34084941 PMCID: PMC8170776 DOI: 10.1212/nxg.0000000000000587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Teerin Liewluck
- Division of Neuromuscular Medicine and Muscle Laboratory, Department of Neurology, Mayo Clinic, Rochester, MN
| |
Collapse
|
19
|
Kley RA, Leber Y, Schrank B, Zhuge H, Orfanos Z, Kostan J, Onipe A, Sellung D, Güttsches AK, Eggers B, Jacobsen F, Kress W, Marcus K, Djinovic-Carugo K, van der Ven PFM, Fürst DO, Vorgerd M. FLNC-Associated Myofibrillar Myopathy: New Clinical, Functional, and Proteomic Data. NEUROLOGY-GENETICS 2021; 7:e590. [PMID: 34235269 PMCID: PMC8237399 DOI: 10.1212/nxg.0000000000000590] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 12/28/2020] [Indexed: 11/15/2022]
Abstract
Objective To determine whether a new indel mutation in the dimerization domain of filamin C (FLNc) causes a hereditary myopathy with protein aggregation in muscle fibers, we clinically and molecularly studied a German family with autosomal dominant myofibrillar myopathy (MFM). Methods We performed mutational analysis in 3 generations, muscle histopathology, and proteomic studies of IM protein aggregates. Functional consequences of the FLNC mutation were investigated with interaction and transfection studies and biophysics molecular analysis. Results Eight patients revealed clinical features of slowly progressive proximal weakness associated with a heterozygous c.8025_8030delCAAGACinsA (p.K2676Pfs*3) mutation in FLNC. Two patients exhibited a mild cardiomyopathy. MRI of skeletal muscle revealed lipomatous changes typical for MFM with FLNC mutations. Muscle biopsies showed characteristic MFM findings with protein aggregation and lesion formation. The proteomic profile of aggregates was specific for MFM-filaminopathy and indicated activation of the ubiquitin-proteasome system (UPS) and autophagic pathways. Functional studies revealed that mutant FLNc is misfolded, unstable, and incapable of forming homodimers and heterodimers with wild-type FLNc. Conclusions This new MFM-filaminopathy family confirms that expression of mutant FLNC leads to an adult-onset muscle phenotype with intracellular protein accumulation. Mutant FLNc protein is biochemically compromised and leads to dysregulation of protein quality control mechanisms. Proteomic analysis of MFM protein aggregates is a potent method to identify disease-relevant proteins, differentiate MFM subtypes, evaluate the relevance of gene variants, and identify novel MFM candidate genes.
Collapse
Affiliation(s)
- Rudolf Andre Kley
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Yvonne Leber
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Bertold Schrank
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Heidi Zhuge
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Zacharias Orfanos
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Julius Kostan
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Adekunle Onipe
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Dominik Sellung
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Anne Katrin Güttsches
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Britta Eggers
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Frank Jacobsen
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Wolfram Kress
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Katrin Marcus
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Kristina Djinovic-Carugo
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Peter F M van der Ven
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Dieter O Fürst
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| | - Matthias Vorgerd
- Department of Neurology (R.A.K., H.Z., D.S., A.K.G., F.J., M.V.), Heimer Institute for Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany; Department of Neurology and Clinical Neurophysiology (R.A.K.), St. Marien-Hospital Borken, Borken, Germany; Department of Molecular Cell Biology (Y.L., Z.O., P.F.M.V., D.O.F.), Institute for Cell Biology, University of Bonn, Bonn, Germany; Department of Neurology (B.S.), DKD HELIOS Klinik Wiesbaden, Wiesbaden, Germany; Department of Structural and Computational Biology (J.K., A.O., K.D.-C.), Max Perutz Laboratories, University of Vienna, Vienna, Austria; Medizinisches Proteom-Center (B.E., K.M.), Ruhr-University Bochum, Bochum, Germany; Institute of Human Genetics (W.K.), University of Würzburg, Würzburg, Germany; and Department of Biochemistry (K.D.-C.), Faculty of Chemistry and Chemical Technology, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
20
|
The Role of Z-disc Proteins in Myopathy and Cardiomyopathy. Int J Mol Sci 2021; 22:ijms22063058. [PMID: 33802723 PMCID: PMC8002584 DOI: 10.3390/ijms22063058] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/07/2021] [Accepted: 03/11/2021] [Indexed: 12/11/2022] Open
Abstract
The Z-disc acts as a protein-rich structure to tether thin filament in the contractile units, the sarcomeres, of striated muscle cells. Proteins found in the Z-disc are integral for maintaining the architecture of the sarcomere. They also enable it to function as a (bio-mechanical) signalling hub. Numerous proteins interact in the Z-disc to facilitate force transduction and intracellular signalling in both cardiac and skeletal muscle. This review will focus on six key Z-disc proteins: α-actinin 2, filamin C, myopalladin, myotilin, telethonin and Z-disc alternatively spliced PDZ-motif (ZASP), which have all been linked to myopathies and cardiomyopathies. We will summarise pathogenic variants identified in the six genes coding for these proteins and look at their involvement in myopathy and cardiomyopathy. Listing the Minor Allele Frequency (MAF) of these variants in the Genome Aggregation Database (GnomAD) version 3.1 will help to critically re-evaluate pathogenicity based on variant frequency in normal population cohorts.
Collapse
|
21
|
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
|
22
|
ten Dam L, de Visser M, Ginjaar IB, van Duyvenvoorde HA, van Koningsbruggen S, van der Kooi AJ. Elucidation of the Genetic Cause in Dutch Limb Girdle Muscular Dystrophy Families: A 27-Year's Journey. J Neuromuscul Dis 2021; 8:261-272. [PMID: 33386810 PMCID: PMC9789482 DOI: 10.3233/jnd-200585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
BACKGROUND A Dutch cohort of 105 carefully selected limb girdle muscular dystrophy (LGMD) patients from 68 families has been subject to genetic testing over the last 20 years. After subsequent targeted gene analysis around two thirds (45/68) of the families had received a genetic diagnosis in 2013. OBJECTIVE To describe the results of further genetic testing in the remaining undiagnosed limb girdle muscular dystrophy families in this cohort. METHODS In the families of the cohort for whom no genetic diagnosis was established (n = 23) further testing using Sanger sequencing, next generation sequencing with gene panel analysis or whole-exome sequencing was performed. In one case DNA analysis for facioscapulohumeral dystrophy type 1 was carried out. RESULTS In eight families no additional genetic tests could be performed. In 12 of the remaining 15 families in which additional testing could be performed a genetic diagnosis was established: two LGMDR1 calpain3-related families with CAPN3 mutations, one LGMDR2 dysferlin-related family with DYSF mutations, three sarcoglycanopathy families (LGMDR3-5 α-, β- and γ-sarcoglycan-related) with SGCA/SGCB/SGCG mutations, one LGMDR8 TRIM 32-related family with TRIM32 mutations, two LGMDR19 GMPPB-related families with GMPPB mutations, one family with MICU1-related myopathy, one family with FLNC-related myopathy and one family with facioscapulohumeral dystrophy type 1. At this moment a genetic diagnosis has been made in 57 of the 60 families of which DNA was available (95%). CONCLUSION A genetic diagnosis is obtained in 95% of the families of the original Dutch LGMD cohort of which DNA was available.
Collapse
Affiliation(s)
- L. ten Dam
- Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands,Correspondence to: Leroy ten Dam, Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Meibergdreef 9, 1105AZ Amsterdam, The Netherlands. Tel.: +31 20 566 9111; E-mail:
| | - M. de Visser
- Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Ieke B. Ginjaar
- Department of Clinical Genetics, Leiden University Medical Centre, Leiden, The Netherlands
| | | | - Silvana van Koningsbruggen
- Department of Clinical Genetics, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam, The Netherlands
| | - Anneke J. van der Kooi
- Department of Neurology, Amsterdam University Medical Centre, University of Amsterdam, Amsterdam Neuroscience, Amsterdam, The Netherlands
| |
Collapse
|
23
|
Knyazeva A, Khudiakov A, Vaz R, Muravyev A, Sukhareva K, Sejersen T, Kostareva A. FLNC Expression Level Influences the Activity of TEAD-YAP/TAZ Signaling. Genes (Basel) 2020; 11:genes11111343. [PMID: 33202721 PMCID: PMC7696573 DOI: 10.3390/genes11111343] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Filamin C (FLNC), being one of the major actin-binding proteins, is involved in the maintenance of key muscle cell functions. Inherited skeletal muscle and cardiac disorders linked to genetic variants in FLNC have attracted attention because of their high clinical importance and possibility of genotype-phenotype correlations. To further expand on the role of FLNC in muscle cells, we focused on detailed alterations of muscle cell properties developed after the loss of FLNC. Using the CRISPR/Cas9 method we generated a C2C12 murine myoblast cell line with stably suppressed Flnc expression. FLNC-deficient myoblasts have a significantly higher proliferation rate combined with an impaired cell migration capacity. The suppression of Flnc expression leads to inability to complete myogenic differentiation, diminished expression of Myh1 and Myh4, alteration of transcriptional dynamics of myogenic factors, such as Mymk and Myog, and deregulation of Hippo signaling pathway. Specifically, we identified elevated basal levels of Hippo activity in myoblasts with loss of FLNC, and ineffective reduction of Hippo signaling activity during myogenic differentiation. The latter was restored by Flnc overexpression. In summary, we confirmed the role of FLNC in muscle cell proliferation, migration and differentiation, and demonstrated for the first time the direct link between Flnc expression and activity of TEAD-YAP\TAZ signaling. These findings support a role of FLNC in regulation of essential muscle processes relying on mechanical as well as signaling mechanisms.
Collapse
Affiliation(s)
- Anastasia Knyazeva
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (A.K.); (A.M.); (K.S.); (A.K.)
- Correspondence:
| | - Aleksandr Khudiakov
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (A.K.); (A.M.); (K.S.); (A.K.)
| | - Raquel Vaz
- Department of Molecular Medicine and Surgery and Center for Molecular Medicine, Karolinska Institute, 171 76 Stockholm, Sweden;
| | - Aleksey Muravyev
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (A.K.); (A.M.); (K.S.); (A.K.)
| | - Ksenia Sukhareva
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (A.K.); (A.M.); (K.S.); (A.K.)
- Graduate School of Life and Health Science, University of Verona, 10 37134 Verona, Italy
| | - Thomas Sejersen
- Department of Women’s and Children’s Health, Karolinska Institute, 171 77 Stockholm, Sweden;
| | - Anna Kostareva
- Almazov National Medical Research Centre, 197341 Saint-Petersburg, Russia; (A.K.); (A.M.); (K.S.); (A.K.)
- Department of Women’s and Children’s Health, Karolinska Institute, 171 77 Stockholm, Sweden;
| |
Collapse
|
24
|
Schuld J, Orfanos Z, Chevessier F, Eggers B, Heil L, Uszkoreit J, Unger A, Kirfel G, van der Ven PFM, Marcus K, Linke WA, Clemen CS, Schröder R, Fürst DO. Homozygous expression of the myofibrillar myopathy-associated p.W2710X filamin C variant reveals major pathomechanisms of sarcomeric lesion formation. Acta Neuropathol Commun 2020; 8:154. [PMID: 32887649 PMCID: PMC7650280 DOI: 10.1186/s40478-020-01001-9] [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: 04/21/2020] [Accepted: 07/22/2020] [Indexed: 01/06/2023] Open
Abstract
Filamin C (FLNc) is mainly expressed in striated muscle cells where it localizes to Z-discs, myotendinous junctions and intercalated discs. Recent studies have revealed numerous mutations in the FLNC gene causing familial and sporadic myopathies and cardiomyopathies with marked clinical variability. The most frequent myopathic mutation, p.W2710X, which is associated with myofibrillar myopathy, deletes the carboxy-terminal 16 amino acids from FLNc and abolishes the dimerization property of Ig-like domain 24. We previously characterized "knock-in" mice heterozygous for this mutation (p.W2711X), and have now investigated homozygous mice using protein and mRNA expression analyses, mass spectrometry, and extensive immunolocalization and ultrastructural studies. Although the latter mice display a relatively mild myopathy under normal conditions, our analyses identified major mechanisms causing the pathophysiology of this disease: in comparison to wildtype animals (i) the expression level of FLNc protein is drastically reduced; (ii) mutant FLNc is relocalized from Z-discs to particularly mechanically strained parts of muscle cells, i.e. myotendinous junctions and myofibrillar lesions; (iii) the number of lesions is greatly increased and these lesions lack Bcl2-associated athanogene 3 (BAG3) protein; (iv) the expression of heat shock protein beta-7 (HSPB7) is almost completely abolished. These findings indicate grave disturbances of BAG3-dependent and -independent autophagy pathways that are required for efficient lesion repair. In addition, our studies reveal general mechanisms of lesion formation and demonstrate that defective FLNc dimerization via its carboxy-terminal domain does not disturb assembly and basic function of myofibrils. An alternative, more amino-terminally located dimerization site might compensate for that loss. Since filamins function as stress sensors, our data further substantiate that FLNc is important for mechanosensing in the context of Z-disc stabilization and maintenance.
Collapse
|
25
|
Mair D, Biskup S, Kress W, Abicht A, Brück W, Zechel S, Knop KC, Koenig FB, Tey S, Nikolin S, Eggermann K, Kurth I, Ferbert A, Weis J. Differential diagnosis of vacuolar myopathies in the NGS era. Brain Pathol 2020; 30:877-896. [PMID: 32419263 PMCID: PMC8017999 DOI: 10.1111/bpa.12864] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Revised: 04/10/2020] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Altered autophagy accompanied by abnormal autophagic (rimmed) vacuoles detectable by light and electron microscopy is a common denominator of many familial and sporadic non-inflammatory muscle diseases. Even in the era of next generation sequencing (NGS), late-onset vacuolar myopathies remain a diagnostic challenge. We identified 32 adult vacuolar myopathy patients from 30 unrelated families, studied their clinical, histopathological and ultrastructural characteristics and performed genetic testing in index patients and relatives using Sanger sequencing and NGS including whole exome sequencing (WES). We established a molecular genetic diagnosis in 17 patients. Pathogenic mutations were found in genes typically linked to vacuolar myopathy (GNE, LDB3/ZASP, MYOT, DES and GAA), but also in genes not regularly associated with severely altered autophagy (FKRP, DYSF, CAV3, COL6A2, GYG1 and TRIM32) and in the digenic facioscapulohumeral muscular dystrophy 2. Characteristic histopathological features including distinct patterns of myofibrillar disarray and evidence of exocytosis proved to be helpful to distinguish causes of vacuolar myopathies. Biopsy validated the pathogenicity of the novel mutations p.(Phe55*) and p.(Arg216*) in GYG1 and of the p.(Leu156Pro) TRIM32 mutation combined with compound heterozygous deletion of exon 2 of TRIM32 and expanded the phenotype of Ala93Thr-caveolinopathy and of limb-girdle muscular dystrophy 2i caused by FKRP mutation. In 15 patients no causal variants were detected by Sanger sequencing and NGS panel analysis. In 12 of these cases, WES was performed, but did not yield any definite mutation or likely candidate gene. In one of these patients with a family history of muscle weakness, the vacuolar myopathy was eventually linked to chloroquine therapy. Our study illustrates the wide phenotypic and genotypic heterogeneity of vacuolar myopathies and validates the role of histopathology in assessing the pathogenicity of novel mutations detected by NGS. In a sizable portion of vacuolar myopathy cases, it remains to be shown whether the cause is hereditary or degenerative.
Collapse
Affiliation(s)
- Dorothea Mair
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany.,Department of Neurology, Kassel School of Medicine, Klinikum Kassel, Kassel, Germany.,University of Southampton, Southampton, UK
| | - Saskia Biskup
- Centre for Genomics and Transcriptomics CeGaT, Tübingen, Germany
| | - Wolfram Kress
- Institute of Human Genetics, University Würzburg, Würzburg, Germany
| | | | - Wolfgang Brück
- Institute of Neuropathology, Göttingen University, Göttingen, Germany
| | - Sabrina Zechel
- Institute of Neuropathology, Göttingen University, Göttingen, Germany
| | | | | | - Shelisa Tey
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| | - Stefan Nikolin
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| | - Katja Eggermann
- Institute of Human Genetics, RWTH Aachen University, Aachen, Germany
| | - Ingo Kurth
- Institute of Human Genetics, RWTH Aachen University, Aachen, Germany
| | - Andreas Ferbert
- Department of Neurology, Kassel School of Medicine, Klinikum Kassel, Kassel, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University, Aachen, Germany
| |
Collapse
|
26
|
Nicolau S, Liewluck T, Milone M. Myopathies with finger flexor weakness: Not only inclusion-body myositis. Muscle Nerve 2020; 62:445-454. [PMID: 32478919 DOI: 10.1002/mus.26914] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2020] [Revised: 04/29/2020] [Accepted: 05/03/2020] [Indexed: 12/11/2022]
Abstract
Muscle disorders are characterized by differential involvement of various muscle groups. Among these, weakness predominantly affecting finger flexors is an uncommon pattern, most frequently found in sporadic inclusion-body myositis. This finding is particularly significant when the full range of histopathological findings of inclusion-body myositis is not found on muscle biopsy. Prominent finger flexor weakness, however, is also observed in other myopathies. It occurs commonly in myotonic dystrophy types 1 and 2. In addition, individual reports and small case series have documented finger flexor weakness in sarcoid and amyloid myopathy, and in inherited myopathies caused by ACTA1, CRYAB, DMD, DYSF, FLNC, GAA, GNE, HNRNPDL, LAMA2, MYH7, and VCP mutations. Therefore, the finding of finger flexor weakness requires consideration of clinical, myopathological, genetic, electrodiagnostic, and sometimes muscle imaging findings to establish a diagnosis.
Collapse
Affiliation(s)
- Stefan Nicolau
- Department of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, Minnesota, 55905, USA
| | - Teerin Liewluck
- Department of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, Minnesota, 55905, USA
| | - Margherita Milone
- Department of Neurology, Mayo Clinic, 200 1st Street SW, Rochester, Minnesota, 55905, USA
| |
Collapse
|
27
|
Reimann L, Schwäble AN, Fricke AL, Mühlhäuser WWD, Leber Y, Lohanadan K, Puchinger MG, Schäuble S, Faessler E, Wiese H, Reichenbach C, Knapp B, Peikert CD, Drepper F, Hahn U, Kreutz C, van der Ven PFM, Radziwill G, Djinović-Carugo K, Fürst DO, Warscheid B. Phosphoproteomics identifies dual-site phosphorylation in an extended basophilic motif regulating FILIP1-mediated degradation of filamin-C. Commun Biol 2020; 3:253. [PMID: 32444788 PMCID: PMC7244511 DOI: 10.1038/s42003-020-0982-5] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/01/2020] [Indexed: 01/10/2023] Open
Abstract
The PI3K/Akt pathway promotes skeletal muscle growth and myogenic differentiation. Although its importance in skeletal muscle biology is well documented, many of its substrates remain to be identified. We here studied PI3K/Akt signaling in contracting skeletal muscle cells by quantitative phosphoproteomics. We identified the extended basophilic phosphosite motif RxRxxp[S/T]xxp[S/T] in various proteins including filamin-C (FLNc). Importantly, this extended motif, located in a unique insert in Ig-like domain 20 of FLNc, is doubly phosphorylated. The protein kinases responsible for this dual-site phosphorylation are Akt and PKCα. Proximity proteomics and interaction analysis identified filamin A-interacting protein 1 (FILIP1) as direct FLNc binding partner. FILIP1 binding induces filamin degradation, thereby negatively regulating its function. Here, dual-site phosphorylation of FLNc not only reduces FILIP1 binding, providing a mechanism to shield FLNc from FILIP1-mediated degradation, but also enables fast dynamics of FLNc necessary for its function as signaling adaptor in cross-striated muscle cells.
Collapse
Affiliation(s)
- Lena Reimann
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Anja N Schwäble
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Anna L Fricke
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Wignand W D Mühlhäuser
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Yvonne Leber
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121, Bonn, Germany
| | - Keerthika Lohanadan
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121, Bonn, Germany
| | - Martin G Puchinger
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, A-1030, Vienna, Austria
| | - Sascha Schäuble
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich-Schiller-University Jena, 07743, Jena, Germany
- Systems Biology and Bioinformatics Unit, Leibniz Institute for Natural Product Research and Infection Biology, Hans Knöll Institute, Jena, Germany
| | - Erik Faessler
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Heike Wiese
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Institute of Pharmacology and Toxicology, University of Ulm, 89081, Ulm, Germany
| | - Christa Reichenbach
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Bettina Knapp
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Christian D Peikert
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Bioinformatics Research & Development, BioNTech SE, 55131, Mainz, Germany
| | - Friedel Drepper
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Udo Hahn
- Jena University Language & Information Engineering (JULIE) Lab, Friedrich-Schiller-University Jena, 07743, Jena, Germany
| | - Clemens Kreutz
- Institute of Medical Biometry and Statistics, Faculty of Medicine and Medical Center, University of Freiburg, 79104, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Peter F M van der Ven
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121, Bonn, Germany
| | - Gerald Radziwill
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Kristina Djinović-Carugo
- Department of Structural and Computational Biology, Max F. Perutz Laboratories, University of Vienna, A-1030, Vienna, Austria
| | - Dieter O Fürst
- Department of Molecular Cell Biology, Institute for Cell Biology, University of Bonn, 53121, Bonn, Germany
| | - Bettina Warscheid
- Biochemistry and Functional Proteomics, Institute of Biology II, Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany.
- Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany.
| |
Collapse
|
28
|
Structure and Function of Filamin C in the Muscle Z-Disc. Int J Mol Sci 2020; 21:ijms21082696. [PMID: 32295012 PMCID: PMC7216277 DOI: 10.3390/ijms21082696] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 04/08/2020] [Accepted: 04/09/2020] [Indexed: 12/22/2022] Open
Abstract
Filamin C (FLNC) is one of three filamin proteins (Filamin A (FLNA), Filamin B (FLNB), and FLNC) that cross-link actin filaments and interact with numerous binding partners. FLNC consists of a N-terminal actin-binding domain followed by 24 immunoglobulin-like repeats with two intervening calpain-sensitive hinges separating R15 and R16 (hinge 1) and R23 and R24 (hinge-2). The FLNC subunit is dimerized through R24 and calpain cleaves off the dimerization domain to regulate mobility of the FLNC subunit. FLNC is localized in the Z-disc due to the unique insertion of 82 amino acid residues in repeat 20 and necessary for normal Z-disc formation that connect sarcomeres. Since phosphorylation of FLNC by PKC diminishes the calpain sensitivity, assembly, and disassembly of the Z-disc may be regulated by phosphorylation of FLNC. Mutations of FLNC result in cardiomyopathy and muscle weakness. Although this review will focus on the current understanding of FLNC structure and functions in muscle, we will also discuss other filamins because they share high sequence similarity and are better characterized. We will also discuss a possible role of FLNC as a mechanosensor during muscle contraction.
Collapse
|
29
|
Verdonschot JAJ, Vanhoutte EK, Claes GRF, Helderman-van den Enden ATJM, Hoeijmakers JGJ, Hellebrekers DMEI, de Haan A, Christiaans I, Lekanne Deprez RH, Boen HM, van Craenenbroeck EM, Loeys BL, Hoedemaekers YM, Marcelis C, Kempers M, Brusse E, van Waning JI, Baas AF, Dooijes D, Asselbergs FW, Barge-Schaapveld DQCM, Koopman P, van den Wijngaard A, Heymans SRB, Krapels IPC, Brunner HG. A mutation update for the FLNC gene in myopathies and cardiomyopathies. Hum Mutat 2020; 41:1091-1111. [PMID: 32112656 PMCID: PMC7318287 DOI: 10.1002/humu.24004] [Citation(s) in RCA: 113] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 02/12/2020] [Accepted: 02/25/2020] [Indexed: 12/11/2022]
Abstract
Filamin C (FLNC) variants are associated with cardiac and muscular phenotypes. Originally, FLNC variants were described in myofibrillar myopathy (MFM) patients. Later, high‐throughput screening in cardiomyopathy cohorts determined a prominent role for FLNC in isolated hypertrophic and dilated cardiomyopathies (HCM and DCM). FLNC variants are now among the more prevalent causes of genetic DCM. FLNC‐associated DCM is associated with a malignant clinical course and a high risk of sudden cardiac death. The clinical spectrum of FLNC suggests different pathomechanisms related to variant types and their location in the gene. The appropriate functioning of FLNC is crucial for structural integrity and cell signaling of the sarcomere. The secondary protein structure of FLNC is critical to ensure this function. Truncating variants with subsequent haploinsufficiency are associated with DCM and cardiac arrhythmias. Interference with the dimerization and folding of the protein leads to aggregate formation detrimental for muscle function, as found in HCM and MFM. Variants associated with HCM are predominantly missense variants, which cluster in the ROD2 domain. This domain is important for binding to the sarcomere and to ensure appropriate cell signaling. We here review FLNC genotype–phenotype correlations based on available evidence.
Collapse
Affiliation(s)
- Job A J Verdonschot
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Cardiology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands
| | - Els K Vanhoutte
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Godelieve R F Claes
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | | | | | - Debby M E I Hellebrekers
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Amber de Haan
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Imke Christiaans
- Department of Clinical Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands.,Department of Clinical Genetics, University Medical Centre Groningen, Groningen, The Netherlands
| | - Ronald H Lekanne Deprez
- Department of Clinical Genetics, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Hanne M Boen
- Department of Cardiology, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium
| | | | - Bart L Loeys
- Department of Medical Genetics, Antwerp University Hospital, University of Antwerp, Antwerp, Belgium
| | - Yvonne M Hoedemaekers
- Department of Clinical Genetics, University Medical Centre Groningen, Groningen, The Netherlands.,Department of Clinical Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Carlo Marcelis
- Department of Clinical Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Marlies Kempers
- Department of Clinical Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Esther Brusse
- Department of Neurology, Erasmus MC University Medical Centre, Rotterdam, The Netherlands
| | - Jaap I van Waning
- Department of Clinical Genetics, Erasmus Medical Center, Rotterdam, The Netherlands.,Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Annette F Baas
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Dennis Dooijes
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Folkert W Asselbergs
- Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
| | | | | | - Arthur van den Wijngaard
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Stephane R B Heymans
- Department of Cardiology, Cardiovascular Research Institute (CARIM), Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Cardiovascular Sciences, Centre for Molecular and Vascular Biology, KU Leuven, Leuven, Belgium.,The Netherlands Heart Institute, Utrecht, The Netherlands
| | - Ingrid P C Krapels
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands
| | - Han G Brunner
- Department of Clinical Genetics, Maastricht University Medical Center, Maastricht, The Netherlands.,Department of Clinical Genetics, Radboud University Medical Centre, Nijmegen, The Netherlands.,Department of Genetics and Cell Biology, GROW Institute for Developmental Biology and Cancer, Maastricht University Medical Centre, Maastricht, The Netherlands
| |
Collapse
|
30
|
Lee HCH, Wong S, Sheng B, Pan NYK, Leung YKF, Lau KKD, Cheng YS, Ho LC, Li R, Lee CN, Tsoi TH, Cheung YFN, Fu YPM, Kan NCA, Chu YP, Au WCL, Yeung HMJ, Li SH, Cheung CFM, Tong HF, Hung LYE, Chan TYC, Li CT, Tong TYT, Tong TWC, Leung HYC, Lee KH, Yeung SYS, Lee SYB, Lau TCG, Lam CW, Mak CM, Chan AYW. Clinical and pathological characterization of FLNC-related myofibrillar myopathy caused by founder variant c.8129G>A in Hong Kong Chinese. Clin Genet 2020; 97:747-757. [PMID: 32022900 DOI: 10.1111/cge.13715] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 01/17/2020] [Accepted: 01/28/2020] [Indexed: 12/18/2022]
Abstract
FLNC-related myofibrillar myopathy could manifest as autosomal dominant late-onset slowly progressive proximal muscle weakness; involvements of cardiac and/or respiratory functions are common. We describe 34 patients in nine families of FLNC-related myofibrillar myopathy in Hong Kong ethnic Chinese diagnosed over the last 12 years, in whom the same pathogenic variant c.8129G>A (p.Trp2710*) was detected. Twenty-six patients were symptomatic when diagnosed; four patients died of pneumonia and/or respiratory failure. Abnormal amorphous material or granulofilamentous masses were detected in half of the cases, with mitochondrial abnormalities noted in two-thirds. We also show by haplotype analysis the founder effect associated with this Hong Kong variant, which might have occurred 42 to 71 generations ago or around Tang and Song dynasties, and underlain a higher incidence of myofibrillar myopathy among Hong Kong Chinese. The late-onset nature and slowly progressive course of the highly penetrant condition could have significant impact on the family members, and an early diagnosis could benefit the whole family. Considering another neighboring founder variant in FLNC in German patients, we advocate development of specific therapies such as chaperone-based or antisense oligonucleotide strategies for this particular type of myopathy.
Collapse
Affiliation(s)
| | - Shun Wong
- Department of Pathology, Princess Margaret Hospital, Hong Kong.,Pathology Department, St. Paul's Hospital, Hong Kong
| | - Bun Sheng
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong
| | - Nin-Yuan Keith Pan
- Department of Diagnostic Radiology, Princess Margaret Hospital, Hong Kong
| | | | | | - Yue Sandy Cheng
- Department of Clinical Pathology, Pamela Youde Nethersole Eastern Hospital, Hong Kong.,Department of Clinical Laboratory, Gleneagles Hong Kong Hospital, Hong Kong
| | - Luen-Cheung Ho
- Department of Pathology, Queen Elizabeth Hospital, Hong Kong
| | - Richard Li
- Department of Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong
| | - Chi-Nam Lee
- Department of Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong
| | - Tak-Hong Tsoi
- Department of Medicine, Pamela Youde Nethersole Eastern Hospital, Hong Kong
| | | | | | | | - Yim-Pui Chu
- Department of Medicine and Geriatrics, Princess Margaret Hospital, Hong Kong
| | - Wing-Chi Lisa Au
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong
| | | | - Siu-Hung Li
- Department of Medicine, North District Hospital, Hong Kong
| | | | - Hok-Fung Tong
- Department of Pathology, Princess Margaret Hospital, Hong Kong
| | | | | | - Chi Terence Li
- Department of Pathology, Princess Margaret Hospital, Hong Kong
| | | | | | | | - Ka-Ho Lee
- Department of Pathology, Princess Margaret Hospital, Hong Kong
| | | | | | | | - Ching-Wan Lam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong
| | - Chloe Miu Mak
- Department of Pathology, Princess Margaret Hospital, Hong Kong
| | | |
Collapse
|
31
|
Xiao F, Wei Q, Wu B, Liu X, Mading A, Yang L, Li Y, Liu F, Pan X, Wang H. Clinical exome sequencing revealed that FLNC variants contribute to the early diagnosis of cardiomyopathies in infant patients. Transl Pediatr 2020; 9:21-33. [PMID: 32154132 PMCID: PMC7036646 DOI: 10.21037/tp.2019.12.02] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND FLNC encodes actin-binding protein and is mainly concentrated in skeletal and cardiac muscle. Mutations in FLNC were found in cardiomyopathies. To date, studies on FLNC-cardiomyopathies have mainly been reported in adults. There are limited studies that have investigated FLNC variants in pediatric patients with cardiomyopathies. METHODS We summarized the patients who carried rare variants of FLNC from May 2016 to May 2019 in the Center for Molecular Medicine, Children's Hospital of Fudan University, from clinical exome sequencing data. RESULTS A total of 5 patients with FLNC rare variants were included. Of them, 3 were male and 2 were female. The median age was 3 months (range from 19 days to 30 months). A1186V was a known pathogenic variant reported in pediatric patients with cardiomyopathy (PMID: 29858533), and the other four variants were novel. In the four novel variants, there are one splicing (c.2265+4del) and three missense (p.R441I, p.C1639Y, and p.A2648S). Two patients (patients 1 and 3) were diagnosed with restrictive cardiomyopathy, two patients (patients 2 and 5) were diagnosed with dilated cardiomyopathy, and one patient (patient 4) was diagnosed with arrhythmia. CONCLUSIONS All five patients have survived to date. In summary, FLNC rare variants identified by clinical exome sequencing provide genetic evidence to make early diagnosis of cardiomyopathy in infant patients.
Collapse
Affiliation(s)
- Feifan Xiao
- Depatment of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China.,Center for Molecular Medicine, Children's Hospital of Fudan University; Institutes of Biomedical Sciences, Fudan University, Shanghai 201102, China
| | - Qiufen Wei
- The Maternal & Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Bingbing Wu
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xu Liu
- Depatment of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Aiyao Mading
- Depatment of Neonatology, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Lin Yang
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Yan Li
- The Maternal & Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Fang Liu
- Cardiovascular center, Children's Hospital of Fudan University, Shanghai 201102, China
| | - Xinnian Pan
- The Maternal & Child Health Hospital of Guangxi Zhuang Autonomous Region, Nanning 530021, China
| | - Huijun Wang
- Center for Molecular Medicine, Children's Hospital of Fudan University, Shanghai 201102, China
| |
Collapse
|
32
|
A mutation in the filamin c gene causes myofibrillar myopathy with lower motor neuron syndrome: a case report. BMC Neurol 2019; 19:198. [PMID: 31421687 PMCID: PMC6697925 DOI: 10.1186/s12883-019-1410-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Accepted: 07/23/2019] [Indexed: 11/13/2022] Open
Abstract
Background Myofibrillar myopathies (MFMs) are a genetically heterogeneous group of muscle disorders. Mutations in the filamin C gene (FLNC) have previously been identified in patients with MFM. The phenotypes of FLNC-related MFM are heterogeneous. Case presentation The patient was a 37-year-old male who first experienced weakness in the distal muscles of his hand, which eventually spread to the lower limbs and proximal muscles. Serum creatine kinase levels were moderately elevated. Obvious neuropathic changes in the electromyographic exam and edema changes in lower distal limb magnetic resonance imaging were observed. Histopathological examination revealed the presence of abnormal protein aggregates and angular atrophy in some muscle fibers. Ultrastructural analysis showed inordinate myofibrillar structures and dissolved myofilaments. DNA sequencing analysis detected a heterozygous missense mutation (c.7123G > A, p.V2375I) in the immunoglobulin (Ig)-like domain 21 of FLNC. Conclusions FLNC mutation c.7123G > A, p.V2375I in the immunoglobulin (Ig)-like domain 21 can be associated with distal myopathy with typical MFM features and lower motor neuron syndrome. Although electromyographic examination of our patient showed obvious neuropathic changes, MFM could not be excluded. Therefore, genetic testing is necessary to make an accurate diagnosis. Electronic supplementary material The online version of this article (10.1186/s12883-019-1410-7) contains supplementary material, which is available to authorized users.
Collapse
|
33
|
Al-Tahan S, Weiss L, Yu H, Tang S, Saporta M, Vihola A, Mozaffar T, Udd B, Kimonis V. New family with HSPB8-associated autosomal dominant rimmed vacuolar myopathy. NEUROLOGY-GENETICS 2019; 5:e349. [PMID: 31403083 PMCID: PMC6659134 DOI: 10.1212/nxg.0000000000000349] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 05/16/2019] [Indexed: 12/29/2022]
Abstract
Objective We clinically and molecularly characterize a new family with autosomal dominant rimmed vacuolar myopathy (RVM) caused by mutations in the HSPB8 gene. Methods We performed whole-exome and whole-genome sequencing in the family. Western blot and immunocytochemistry were used to analyze 3 patient fibroblasts, and findings were compared with their age- and sex-matched controls. Results Affected patients have distal and proximal myopathy, with muscle biopsy showing rimmed vacuoles, muscle fiber atrophy, and endomysial fibrosis typical of RVM. Muscle MRI showed severe relatively symmetric multifocal fatty degenerative changes of the lower extremities. We identified a duplication of C at position 515 of the HSPB8 gene (c.515dupC) by whole-genome sequencing, which caused a frameshift with a predicted alternate stop codon p.P173SFS*43 in all affected individuals, resulting in an elongated protein product. Western blot and immunocytochemistry studies revealed reduced expression of heat shock protein beta 8 in patient fibroblasts compared with control fibroblasts, in addition to disrupted autophagy pathology. Conclusions We report a novel family with autosomal dominant RVM caused by the c.515dupC mutation of the HSPB8 gene, causing a translational frameshift that results in an elongated protein. Understanding the mechanism for the RVM pathology caused by mutated chaperone will permit novel targeted strategies to alter the natural history progression. As next-generation sequencing becomes more available, additional myopathic families will be identified with HSPB8 mutations.
Collapse
Affiliation(s)
- Sejad Al-Tahan
- Division of Genetics and Genomic Medicine (S.A.-T., L.W., H.Y.), Department of Pediatrics, University of California, Irvine; Opti West (S.A.-T.), West Anaheim Medical Center, Anaheim; Ambry Genetics (S.T.), Mission Viejo, CA; Miller School of Medicine (M.S.), University of Miami, FL; Folkhälsan Institute of Genetics and the Department of Medical Genetics (A.V., B.U.), Medicum, University of Helsinki; Neuromuscular Research Center (A.V., B.U.), Tampere University and University Hospital, Neurology, Finland; Neuromuscular Program (T.M.), Department of Neurology, University of California-Irvine, Orange; and Neurology Department (B.U.), Vasa Central Hospital, Finland
| | - Lan Weiss
- Division of Genetics and Genomic Medicine (S.A.-T., L.W., H.Y.), Department of Pediatrics, University of California, Irvine; Opti West (S.A.-T.), West Anaheim Medical Center, Anaheim; Ambry Genetics (S.T.), Mission Viejo, CA; Miller School of Medicine (M.S.), University of Miami, FL; Folkhälsan Institute of Genetics and the Department of Medical Genetics (A.V., B.U.), Medicum, University of Helsinki; Neuromuscular Research Center (A.V., B.U.), Tampere University and University Hospital, Neurology, Finland; Neuromuscular Program (T.M.), Department of Neurology, University of California-Irvine, Orange; and Neurology Department (B.U.), Vasa Central Hospital, Finland
| | - Howard Yu
- Division of Genetics and Genomic Medicine (S.A.-T., L.W., H.Y.), Department of Pediatrics, University of California, Irvine; Opti West (S.A.-T.), West Anaheim Medical Center, Anaheim; Ambry Genetics (S.T.), Mission Viejo, CA; Miller School of Medicine (M.S.), University of Miami, FL; Folkhälsan Institute of Genetics and the Department of Medical Genetics (A.V., B.U.), Medicum, University of Helsinki; Neuromuscular Research Center (A.V., B.U.), Tampere University and University Hospital, Neurology, Finland; Neuromuscular Program (T.M.), Department of Neurology, University of California-Irvine, Orange; and Neurology Department (B.U.), Vasa Central Hospital, Finland
| | - Sha Tang
- Division of Genetics and Genomic Medicine (S.A.-T., L.W., H.Y.), Department of Pediatrics, University of California, Irvine; Opti West (S.A.-T.), West Anaheim Medical Center, Anaheim; Ambry Genetics (S.T.), Mission Viejo, CA; Miller School of Medicine (M.S.), University of Miami, FL; Folkhälsan Institute of Genetics and the Department of Medical Genetics (A.V., B.U.), Medicum, University of Helsinki; Neuromuscular Research Center (A.V., B.U.), Tampere University and University Hospital, Neurology, Finland; Neuromuscular Program (T.M.), Department of Neurology, University of California-Irvine, Orange; and Neurology Department (B.U.), Vasa Central Hospital, Finland
| | - Mario Saporta
- Division of Genetics and Genomic Medicine (S.A.-T., L.W., H.Y.), Department of Pediatrics, University of California, Irvine; Opti West (S.A.-T.), West Anaheim Medical Center, Anaheim; Ambry Genetics (S.T.), Mission Viejo, CA; Miller School of Medicine (M.S.), University of Miami, FL; Folkhälsan Institute of Genetics and the Department of Medical Genetics (A.V., B.U.), Medicum, University of Helsinki; Neuromuscular Research Center (A.V., B.U.), Tampere University and University Hospital, Neurology, Finland; Neuromuscular Program (T.M.), Department of Neurology, University of California-Irvine, Orange; and Neurology Department (B.U.), Vasa Central Hospital, Finland
| | - Anna Vihola
- Division of Genetics and Genomic Medicine (S.A.-T., L.W., H.Y.), Department of Pediatrics, University of California, Irvine; Opti West (S.A.-T.), West Anaheim Medical Center, Anaheim; Ambry Genetics (S.T.), Mission Viejo, CA; Miller School of Medicine (M.S.), University of Miami, FL; Folkhälsan Institute of Genetics and the Department of Medical Genetics (A.V., B.U.), Medicum, University of Helsinki; Neuromuscular Research Center (A.V., B.U.), Tampere University and University Hospital, Neurology, Finland; Neuromuscular Program (T.M.), Department of Neurology, University of California-Irvine, Orange; and Neurology Department (B.U.), Vasa Central Hospital, Finland
| | - Tahseen Mozaffar
- Division of Genetics and Genomic Medicine (S.A.-T., L.W., H.Y.), Department of Pediatrics, University of California, Irvine; Opti West (S.A.-T.), West Anaheim Medical Center, Anaheim; Ambry Genetics (S.T.), Mission Viejo, CA; Miller School of Medicine (M.S.), University of Miami, FL; Folkhälsan Institute of Genetics and the Department of Medical Genetics (A.V., B.U.), Medicum, University of Helsinki; Neuromuscular Research Center (A.V., B.U.), Tampere University and University Hospital, Neurology, Finland; Neuromuscular Program (T.M.), Department of Neurology, University of California-Irvine, Orange; and Neurology Department (B.U.), Vasa Central Hospital, Finland
| | - Bjarne Udd
- Division of Genetics and Genomic Medicine (S.A.-T., L.W., H.Y.), Department of Pediatrics, University of California, Irvine; Opti West (S.A.-T.), West Anaheim Medical Center, Anaheim; Ambry Genetics (S.T.), Mission Viejo, CA; Miller School of Medicine (M.S.), University of Miami, FL; Folkhälsan Institute of Genetics and the Department of Medical Genetics (A.V., B.U.), Medicum, University of Helsinki; Neuromuscular Research Center (A.V., B.U.), Tampere University and University Hospital, Neurology, Finland; Neuromuscular Program (T.M.), Department of Neurology, University of California-Irvine, Orange; and Neurology Department (B.U.), Vasa Central Hospital, Finland
| | - Virginia Kimonis
- Division of Genetics and Genomic Medicine (S.A.-T., L.W., H.Y.), Department of Pediatrics, University of California, Irvine; Opti West (S.A.-T.), West Anaheim Medical Center, Anaheim; Ambry Genetics (S.T.), Mission Viejo, CA; Miller School of Medicine (M.S.), University of Miami, FL; Folkhälsan Institute of Genetics and the Department of Medical Genetics (A.V., B.U.), Medicum, University of Helsinki; Neuromuscular Research Center (A.V., B.U.), Tampere University and University Hospital, Neurology, Finland; Neuromuscular Program (T.M.), Department of Neurology, University of California-Irvine, Orange; and Neurology Department (B.U.), Vasa Central Hospital, Finland
| |
Collapse
|
34
|
Borch JDS, Eisum ASV, Krag T, Vissing J. Expanding the phenotype of filamin-C-related myofibrillar myopathy. Clin Neurol Neurosurg 2019; 176:30-33. [DOI: 10.1016/j.clineuro.2018.11.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 11/01/2018] [Accepted: 11/17/2018] [Indexed: 02/08/2023]
|
35
|
Schubert J, Tariq M, Geddes G, Kindel S, Miller EM, Ware SM. Novel pathogenic variants in filamin C identified in pediatric restrictive cardiomyopathy. Hum Mutat 2018; 39:2083-2096. [DOI: 10.1002/humu.23661] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2018] [Revised: 08/29/2018] [Accepted: 09/25/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Jeffrey Schubert
- Department of Molecular Genetics, Microbiology, and Biochemistry; University of Cincinnati College of Medicine; Cincinnati Ohio
- Departments of Pediatrics and Medical and Molecular Genetics; Indiana University School of Medicine; Indianapolis Indiana
| | - Muhammad Tariq
- Faculty of Applied Medical Science; University of Tabuk; Tabuk Kingdom of Saudi Arabia
| | - Gabrielle Geddes
- Department of Pediatrics; Medical College of Wisconsin; Milwaukee Wisconsin
| | - Steven Kindel
- Department of Pediatrics; Medical College of Wisconsin; Milwaukee Wisconsin
| | - Erin M. Miller
- Cincinnati Children's Hospital Medical Center; Cincinnati Ohio
| | - Stephanie M. Ware
- Departments of Pediatrics and Medical and Molecular Genetics; Indiana University School of Medicine; Indianapolis Indiana
| |
Collapse
|
36
|
Kiselev A, Vaz R, Knyazeva A, Khudiakov A, Tarnovskaya S, Liu J, Sergushichev A, Kazakov S, Frishman D, Smolina N, Pervunina T, Jorholt J, Sjoberg G, Vershinina T, Rudenko D, Arner A, Sejersen T, Lindstrand A, Kostareva A. De novo mutations in FLNC
leading to early-onset restrictive cardiomyopathy and congenital myopathy. Hum Mutat 2018; 39:1161-1172. [DOI: 10.1002/humu.23559] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 05/22/2018] [Accepted: 05/30/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Artem Kiselev
- Almazov National Medical Research Centre; Saint Petersburg Russia
| | - Raquel Vaz
- Department of Molecular Medicine and Surgery and Center for molecular medicine; Karolinska Institutet; Stockholm Sweden
| | | | | | - Svetlana Tarnovskaya
- Almazov National Medical Research Centre; Saint Petersburg Russia
- Peter the Great St.Petersburg Polytechnic University; Saint Petersburg Russia
| | - Jiao Liu
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| | | | | | - Dmitrij Frishman
- Peter the Great St.Petersburg Polytechnic University; Saint Petersburg Russia
- Department of Bioinformatics; Technische Universität München; Wissenschaftszentrum Weihenstephan; Freising Germany
| | - Natalia Smolina
- Almazov National Medical Research Centre; Saint Petersburg Russia
- ITMO University; Saint Petersburg Russia
- Department of Women's and Children's Health and Center for Molecular Medicine; Karolinska Institute; Stockholm Sweden
| | | | - John Jorholt
- Department of Women's and Children's Health and Center for Molecular Medicine; Karolinska Institute; Stockholm Sweden
| | - Gunnar Sjoberg
- Department of Women's and Children's Health and Center for Molecular Medicine; Karolinska Institute; Stockholm Sweden
| | | | | | - Anders Arner
- Department of Physiology and Pharmacology; Karolinska Institutet; Stockholm Sweden
| | - Thomas Sejersen
- Department of Women's and Children's Health and Center for Molecular Medicine; Karolinska Institute; Stockholm Sweden
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery and Center for molecular medicine; Karolinska Institutet; Stockholm Sweden
- Clinical Genetics; Karolinska University Laboratory; Karolinska University Hospital; Stockholm Sweden
| | - Anna Kostareva
- Almazov National Medical Research Centre; Saint Petersburg Russia
- Department of Women's and Children's Health and Center for Molecular Medicine; Karolinska Institute; Stockholm Sweden
| |
Collapse
|
37
|
Miao J, Su FF, Liu XM, Wei XJ, Yuan Y, Yu XF. A case report: a heterozygous deletion (2791_2805 del) in exon 18 of the filamin C gene causing filamin C-related myofibrillar myopathies in a Chinese family. BMC Neurol 2018; 18:79. [PMID: 29866061 PMCID: PMC5985593 DOI: 10.1186/s12883-018-1078-4] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Accepted: 05/22/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Filamin C-related myofibrillar myopathies (MFM) are progressive skeletal myopathies with an autosomal dominant inheritance pattern. The conditions are caused by mutations of the filamin C gene (FLNC) located in the chromosome 7q32-q35 region. Genetic variations in the FLNC gene result in various clinical phenotypes. CASE PRESENTATION We describe a 43-year-old woman who suffered filamin C-related MFM, with symptoms first presenting in the proximal muscles of the lower limbs and eventually spreading to the upper limbs and distal muscles. The patient's serum level of creatine kinase was mildly increased. Mildy myopathic changes in the electromyographic exam and moderate lipomatous alterations in lower limb MRI were found. Histopathological examination revealed increased muscle fiber size variability, disturbances in oxidative enzyme activity, and the presence of abnormal protein aggregates and vacuoles in some muscle fibers. Ultrastructural analysis showed inclusions composed of thin filaments and interspersed granular densities. DNA sequencing analysis detected a novel 15-nucleotide deletion (c.2791_2805del, p.931_935del) in the FLNC gene. The patient's father, sister, brother, three paternal aunts, one paternal uncle, and the uncle's son also had slowly progressive muscle weakness, and thus, we detected an autosomal dominant inheritance pattern of the disorder. CONCLUSIONS A novel heterogeneous 15-nucleotide deletion (c.2791_2805del, p.931_935del) in the Ig-like domain 7 of the FLNC gene was found to cause filamin C-related MFM. This deletion in the FLNC gene causes protein aggregation, abnormalities in muscle structure, and impairment in muscle fiber function, which leads to muscle weakness.
Collapse
Affiliation(s)
- Jing Miao
- Department of Neurology and Neuroscience Center, First Affiliated Hospital of Jilin University, Changchun, 130021 Jilin, People’s Republic of China
| | - Fei-fei Su
- Department of Neurology and Neuroscience Center, First Affiliated Hospital of Jilin University, Changchun, 130021 Jilin, People’s Republic of China
| | - Xue-mei Liu
- Department of Neurology and Neuroscience Center, First Affiliated Hospital of Jilin University, Changchun, 130021 Jilin, People’s Republic of China
| | - Xiao-jing Wei
- Department of Neurology and Neuroscience Center, First Affiliated Hospital of Jilin University, Changchun, 130021 Jilin, People’s Republic of China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, #8 Xishiku St, Xicheng District, Beijing, 100034 China
| | - Xue-fan Yu
- Department of Neurology and Neuroscience Center First Affiliated Hospital of Jilin University, Changchun, 130021 Jilin, People’s Republic of China
| |
Collapse
|
38
|
Begay RL, Graw SL, Sinagra G, Asimaki A, Rowland TJ, Slavov DB, Gowan K, Jones KL, Brun F, Merlo M, Miani D, Sweet M, Devaraj K, Wartchow EP, Gigli M, Puggia I, Salcedo EE, Garrity DM, Ambardekar AV, Buttrick P, Reece TB, Bristow MR, Saffitz JE, Mestroni L, Taylor MRG. Filamin C Truncation Mutations Are Associated With Arrhythmogenic Dilated Cardiomyopathy and Changes in the Cell-Cell Adhesion Structures. JACC Clin Electrophysiol 2018; 4:504-514. [PMID: 30067491 PMCID: PMC6074050 DOI: 10.1016/j.jacep.2017.12.003] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Revised: 11/20/2017] [Accepted: 12/07/2017] [Indexed: 12/15/2022]
Abstract
OBJECTIVES The purpose of this study was to assess the phenotype of Filamin C (FLNC) truncating variants in dilated cardiomyopathy (DCM) and understand the mechanism leading to an arrhythmogenic phenotype. BACKGROUND Mutations in FLNC are known to lead to skeletal myopathies, which may have an associated cardiac component. Recently, the clinical spectrum of FLNC mutations has been recognized to include a cardiac-restricted presentation in the absence of skeletal muscle involvement. METHODS A population of 319 U.S. and European DCM cardiomyopathy families was evaluated using whole-exome and targeted next-generation sequencing. FLNC truncation probands were identified and evaluated by clinical examination, histology, transmission electron microscopy, and immunohistochemistry. RESULTS A total of 13 individuals in 7 families (2.2%) were found to harbor 6 different FLNC truncation variants (2 stopgain, 1 frameshift, and 3 splicing). Of the 13 FLNC truncation carriers, 11 (85%) had either ventricular arrhythmias or sudden cardiac death, and 5 (38%) presented with evidence of right ventricular dilation. Pathology analysis of 2 explanted hearts from affected FLNC truncation carriers showed interstitial fibrosis in the right ventricle and epicardial fibrofatty infiltration in the left ventricle. Ultrastructural findings included occasional disarray of Z-discs within the sarcomere. Immunohistochemistry showed normal plakoglobin signal at cell-cell junctions, but decreased signals for desmoplakin and synapse-associated protein 97 in the myocardium and buccal mucosa. CONCLUSIONS We found FLNC truncating variants, present in 2.2% of DCM families, to be associated with a cardiac-restricted arrhythmogenic DCM phenotype characterized by a high risk of life-threatening ventricular arrhythmias and a pathological cellular phenotype partially overlapping with arrhythmogenic right ventricular cardiomyopathy.
Collapse
Affiliation(s)
- Rene L Begay
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado
| | - Sharon L Graw
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado
| | - Gianfranco Sinagra
- Department of Cardiology, Ospedali Riuniti and University of Trieste, Trieste, Italy
| | - Angeliki Asimaki
- Department of Pathology, Beth Israel Deaconess Medical Center & Harvard Medical School, Boston, Massachusetts
| | - Teisha J Rowland
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado
| | - Dobromir B Slavov
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado
| | - Katherine Gowan
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado Denver, Aurora, Colorado
| | - Kenneth L Jones
- Department of Pediatrics, Section of Hematology, Oncology, and Bone Marrow Transplant, University of Colorado Denver, Aurora, Colorado
| | - Francesca Brun
- Department of Cardiology, Ospedali Riuniti and University of Trieste, Trieste, Italy
| | - Marco Merlo
- Department of Cardiology, Ospedali Riuniti and University of Trieste, Trieste, Italy
| | - Daniela Miani
- Department of Cardiothoracic Science, University Hospital S. Maria della Misericordia, Udine, Italy
| | - Mary Sweet
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado
| | - Kalpana Devaraj
- Department of Pathology, University of Colorado, University Hospital, Aurora, Colorado
| | - Eric P Wartchow
- Department of Pathology, Children's Hospital Colorado, Aurora, Colorado
| | - Marta Gigli
- Department of Cardiology, Ospedali Riuniti and University of Trieste, Trieste, Italy
| | - Ilaria Puggia
- Department of Cardiology, Ospedali Riuniti and University of Trieste, Trieste, Italy
| | - Ernesto E Salcedo
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado
| | - Deborah M Garrity
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, Colorado
| | - Amrut V Ambardekar
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado
| | - Peter Buttrick
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado
| | - T Brett Reece
- Department of Surgery, University of Colorado Denver, Aurora, Colorado
| | - Michael R Bristow
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado
| | - Jeffrey E Saffitz
- Department of Pathology, Beth Israel Deaconess Medical Center & Harvard Medical School, Boston, Massachusetts
| | - Luisa Mestroni
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado
| | - Matthew R G Taylor
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, Colorado.
| |
Collapse
|
39
|
Castets P, Frank S, Sinnreich M, Rüegg MA. "Get the Balance Right": Pathological Significance of Autophagy Perturbation in Neuromuscular Disorders. J Neuromuscul Dis 2018; 3:127-155. [PMID: 27854220 PMCID: PMC5271579 DOI: 10.3233/jnd-160153] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Recent research has revealed that autophagy, a major catabolic process in cells, is dysregulated in several neuromuscular diseases and contributes to the muscle wasting caused by non-muscle disorders (e.g. cancer cachexia) or during aging (i.e. sarcopenia). From there, the idea arose to interfere with autophagy or manipulate its regulatory signalling to help restore muscle homeostasis and attenuate disease progression. The major difficulty for the development of therapeutic strategies is to restore a balanced autophagic flux, due to the dynamic nature of autophagy. Thus, it is essential to better understand the mechanisms and identify the signalling pathways at play in the control of autophagy in skeletal muscle. A comprehensive analysis of the autophagic flux and of the causes of its dysregulation is required to assess the pathogenic role of autophagy in diseased muscle. Furthermore, it is essential that experiments distinguish between primary dysregulation of autophagy (prior to disease onset) and impairments as a consequence of the pathology. Of note, in most muscle disorders, autophagy perturbation is not caused by genetic modification of an autophagy-related protein, but rather through indirect alteration of regulatory signalling or lysosomal function. In this review, we will present the mechanisms involved in autophagy, and those ensuring its tight regulation in skeletal muscle. We will then discuss as to how autophagy dysregulation contributes to the pathogenesis of neuromuscular disorders and possible ways to interfere with this process to limit disease progression.
Collapse
Affiliation(s)
| | - Stephan Frank
- Institute of Pathology, Division of Neuropathology Basel University Hospital, Basel, Switzerland
| | - Michael Sinnreich
- Neuromuscular Research Center, Departments of Neurology and Biomedicine, Pharmazentrum, Basel, Switzerland
| | | |
Collapse
|
40
|
Tasca G, Udd B. Hereditary myopathy with early respiratory failure (HMERF): Still rare, but common enough. Neuromuscul Disord 2018; 28:268-276. [DOI: 10.1016/j.nmd.2017.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/04/2017] [Accepted: 12/03/2017] [Indexed: 01/04/2023]
|
41
|
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: 41] [Impact Index Per Article: 5.9] [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
|
42
|
Rossi D, Palmio J, Evilä A, Galli L, Barone V, Caldwell TA, Policke RA, Aldkheil E, Berndsen CE, Wright NT, Malfatti E, Brochier G, Pierantozzi E, Jordanova A, Guergueltcheva V, Romero NB, Hackman P, Eymard B, Udd B, Sorrentino V. A novel FLNC frameshift and an OBSCN variant in a family with distal muscular dystrophy. PLoS One 2017; 12:e0186642. [PMID: 29073160 PMCID: PMC5657976 DOI: 10.1371/journal.pone.0186642] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 10/04/2017] [Indexed: 11/30/2022] Open
Abstract
A novel FLNC c.5161delG (p.Gly1722ValfsTer61) mutation was identified in two members of a French family affected by distal myopathy and in one healthy relative. This FLNC c.5161delG mutation is one nucleotide away from a previously reported FLNC mutation (c.5160delC) that was identified in patients and in asymptomatic carriers of three Bulgarian families with distal muscular dystrophy, indicating a low penetrance of the FLNC frameshift mutations. Given these similarities, we believe that the two FLNC mutations alone can be causative of distal myopathy without full penetrance. Moreover, comparative analysis of the clinical manifestations indicates that patients of the French family show an earlier onset and a complete segregation of the disease. As a possible explanation of this, the two French patients also carry a OBSCN c.13330C>T (p.Arg4444Trp) mutation. The p.Arg4444Trp variant is localized within the OBSCN Ig59 domain that, together with Ig58, binds to the ZIg9/ZIg10 domains of titin at Z-disks. Structural and functional studies indicate that this OBSCN p.Arg4444Trp mutation decreases titin binding by ~15-fold. On this line, we suggest that the combination of the OBSCN p.Arg4444Trp variant and of the FLNC c.5161delG mutation, can cooperatively affect myofibril stability and increase the penetrance of muscular dystrophy in the French family.
Collapse
Affiliation(s)
- Daniela Rossi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena and Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Johanna Palmio
- Neuromuscular Research Center, Tampere University and University Hospital, Tampere, Finland
| | - Anni Evilä
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Lucia Galli
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena and Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Virginia Barone
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena and Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Tracy A. Caldwell
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, United States of America
| | - Rachel A. Policke
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, United States of America
| | - Esraa Aldkheil
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, United States of America
| | - Christopher E. Berndsen
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, United States of America
| | - Nathan T. Wright
- Department of Chemistry and Biochemistry, James Madison University, Harrisonburg, Virginia, United States of America
| | - Edoardo Malfatti
- Neuromuscular Morphology Unit, and Reference Center for Neuromuscular Diseases, Myology Institute, Groupe Hospitalier La Pitié-Salpêtrière, Paris, France
| | - Guy Brochier
- Neuromuscular Morphology Unit, and Reference Center for Neuromuscular Diseases, Myology Institute, Groupe Hospitalier La Pitié-Salpêtrière, Paris, France
| | - Enrico Pierantozzi
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena and Azienda Ospedaliera Universitaria Senese, Siena, Italy
| | - Albena Jordanova
- Molecular Neurogenomics Group, University of Antwerp, Antwerp, Belgium
- Molecular Medicine Center, Department of Medical Chemistry and Biochemistry, Medical University-Sofia, Sofia, Bulgaria
| | | | - Norma Beatriz Romero
- Neuromuscular Morphology Unit, and Reference Center for Neuromuscular Diseases, Myology Institute, Groupe Hospitalier La Pitié-Salpêtrière, Paris, France
| | - Peter Hackman
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Bruno Eymard
- Neuromuscular Morphology Unit, and Reference Center for Neuromuscular Diseases, Myology Institute, Groupe Hospitalier La Pitié-Salpêtrière, Paris, France
| | - Bjarne Udd
- Neuromuscular Research Center, Tampere University and University Hospital, Tampere, Finland
- Folkhälsan Institute of Genetics and Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
- Department of Neurology, Vaasa Central Hospital, Vaasa, Finland
| | - Vincenzo Sorrentino
- Molecular Medicine Section, Department of Molecular and Developmental Medicine, University of Siena and Azienda Ospedaliera Universitaria Senese, Siena, Italy
- * E-mail:
| |
Collapse
|
43
|
Unger A, Beckendorf L, Böhme P, Kley R, von Frieling-Salewsky M, Lochmüller H, Schröder R, Fürst DO, Vorgerd M, Linke WA. Translocation of molecular chaperones to the titin springs is common in skeletal myopathy patients and affects sarcomere function. Acta Neuropathol Commun 2017; 5:72. [PMID: 28915917 PMCID: PMC5603016 DOI: 10.1186/s40478-017-0474-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2017] [Accepted: 09/09/2017] [Indexed: 12/16/2022] Open
Abstract
Myopathies encompass a wide variety of acquired and hereditary disorders. The pathomechanisms include structural and functional changes affecting, e.g., myofiber metabolism and contractile properties. In this study, we observed increased passive tension (PT) of skinned myofibers from patients with myofibrillar myopathy (MFM) caused by FLNC mutations (MFM-filaminopathy) and limb-girdle muscular dystrophy type-2A due to CAPN3 mutations (LGMD2A), compared to healthy control myofibers. Because the giant protein titin determines myofiber PT, we measured its molecular size and the titin-to-myosin ratio, but found no differences between myopathies and controls. All-titin phosphorylation and site-specific phosphorylation in the PEVK region were reduced in myopathy, which would be predicted to lower PT. Electron microscopy revealed extensive ultrastructural changes in myofibers of various hereditary myopathies and also suggested massive binding of proteins to the sarcomeric I-band region, presumably heat shock proteins (HSPs), which can translocate to elastic titin under stress conditions. Correlative immunofluorescence and immunoelectron microscopy showed that two small HSPs (HSP27 and αB-crystallin) and the ATP-dependent chaperone HSP90 translocated to the titin springs in myopathy. The small HSPs, but not HSP90, were upregulated in myopathic versus control muscles. The titin-binding pattern of chaperones was regularly observed in Duchenne muscular dystrophy (DMD), LGMD2A, MFM-filaminopathy, MFM-myotilinopathy, titinopathy, and inclusion body myopathy due to mutations in valosin-containing protein, but not in acquired sporadic inclusion body myositis. The three HSPs also associated with elastic titin in mouse models of DMD and MFM-filaminopathy. Mechanical measurements on skinned human myofibers incubated with exogenous small HSPs suggested that the elevated PT seen in myopathy is caused, in part, by chaperone-binding to the titin springs. Whereas this interaction may be protective in that it prevents sarcomeric protein aggregation, it also has detrimental effects on sarcomere function. Thus, we identified a novel pathological phenomenon common to many hereditary muscle disorders, which involves sarcomeric alterations.
Collapse
|
44
|
Papizan JB, Garry GA, Brezprozvannaya S, McAnally JR, Bassel-Duby R, Liu N, Olson EN. Deficiency in Kelch protein Klhl31 causes congenital myopathy in mice. J Clin Invest 2017; 127:3730-3740. [PMID: 28872460 DOI: 10.1172/jci93445] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Accepted: 07/19/2017] [Indexed: 01/19/2023] Open
Abstract
Maintenance of muscle structure and function depends on the precise organization of contractile proteins into sarcomeres and coupling of the contractile apparatus to the sarcoplasmic reticulum (SR), which serves as the reservoir for calcium required for contraction. Several members of the Kelch superfamily of proteins, which modulate protein stability as substrate-specific adaptors for ubiquitination, have been implicated in sarcomere formation. The Kelch protein Klhl31 is expressed in a muscle-specific manner under control of the transcription factor MEF2. To explore its functions in vivo, we created a mouse model of Klhl31 loss of function using the CRISPR-Cas9 system. Mice lacking Klhl31 exhibited stunted postnatal skeletal muscle growth, centronuclear myopathy, central cores, Z-disc streaming, and SR dilation. We used proteomics to identify several candidate Klhl31 substrates, including Filamin-C (FlnC). In the Klhl31-knockout mice, FlnC protein levels were highly upregulated with no change in transcription, and we further demonstrated that Klhl31 targets FlnC for ubiquitination and degradation. These findings highlight a role for Klhl31 in the maintenance of skeletal muscle structure and provide insight into the mechanisms underlying congenital myopathies.
Collapse
|
45
|
Ayyadevara S, Balasubramaniam M, Suri P, Mackintosh SG, Tackett AJ, Sullivan DH, Shmookler Reis RJ, Dennis RA. Proteins that accumulate with age in human skeletal-muscle aggregates contribute to declines in muscle mass and function in Caenorhabditis elegans. Aging (Albany NY) 2016; 8:3486-3497. [PMID: 27992858 PMCID: PMC5270681 DOI: 10.18632/aging.101141] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 12/03/2016] [Indexed: 06/06/2023]
Abstract
Protein aggregation increases with age in normal tissues, and with pathology and age in Alzheimer's hippocampus and mouse cardiac muscle. We now ask whether human skeletal muscle accumulates aggregates with age. Detergent-insoluble protein aggregates were isolated from vastus lateralis biopsies from 5 young (23–27 years of age) and 5 older (64-80 years) adults. Aggregates, quantified after gel electrophoresis, contain 2.1-fold more protein (P<0.0001) when isolated from older subjects relative to young. Of 515 proteins identified by liquid chromatography coupled to tandem mass spectrometry, 56 (11%) were significantly more abundant in older muscle, while 21 (4%) were depleted with age (each P<0.05). Orthologs to seven of these proteins were then targeted in C. elegans by RNA interference. Six of the seven knockdown treatments decreased protein aggregation (range 6-45%, P<0.01 to <0.0001) and increased muscle mass (range 1.5- to 1.85-fold, P<0.01 to <0.0001) in aged nematodes, and rescued mobility (range 1.4 to 1.65-fold, P≤0.0005 each) in a nematode amyloidopathy model. We conclude that specific aggregate proteins, discovered as differentially abundant in aging human muscle, have orthologs that contribute functionally to aggregation and age-associated muscle loss in nematodes, and thus can be considered potential drug targets for sarcopenia in humans.
Collapse
Affiliation(s)
- Srinivas Ayyadevara
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
- Reynolds Institute on Aging, Dept. of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Meenakshisundaram Balasubramaniam
- Reynolds Institute on Aging, Dept. of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- BioInformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Pooja Suri
- Reynolds Institute on Aging, Dept. of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Samuel G. Mackintosh
- Department of Biochemistry & Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Alan J. Tackett
- Department of Biochemistry & Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | - Dennis H. Sullivan
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
- Reynolds Institute on Aging, Dept. of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Geriatric Research, Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Robert J. Shmookler Reis
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
- Reynolds Institute on Aging, Dept. of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- BioInformatics Program, University of Arkansas at Little Rock and University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Department of Biochemistry & Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Geriatric Research, Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| | - Richard A. Dennis
- Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
- Reynolds Institute on Aging, Dept. of Geriatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
- Geriatric Research, Education and Clinical Center, Central Arkansas Veterans Healthcare System, Little Rock, AR 72205, USA
| |
Collapse
|
46
|
van den Bogaart FJA, Claeys KG, Kley RA, Kusters B, Schrading S, Kamsteeg EJ, Voermans NC. Widening the spectrum of filamin-C myopathy: Predominantly proximal myopathy due to the p.A193T mutation in the actin-binding domain of FLNC. Neuromuscul Disord 2016; 27:73-77. [PMID: 27816332 DOI: 10.1016/j.nmd.2016.09.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Revised: 09/13/2016] [Accepted: 09/20/2016] [Indexed: 12/13/2022]
Abstract
We report three patients with a predominantly proximal myopathy due to p.A193T mutation in the actin-binding domain of FLNC, which has so far only been associated with a distal myopathy. They presented with a late onset myopathy characterized by predominant limb-girdle and proximal weakness. We describe the clinical, electrophysiological, pathological, muscle imaging and genetic features. One of our patients did not have typical histological features for a myofibrillar myopathy in muscle biopsy. This observation is important for the recognition of the full clinical spectrum of filamin-C-related myopathies. Muscle imaging has an important role in distinguishing the different filamin-C myopathy types.
Collapse
Affiliation(s)
| | - Kristl G Claeys
- Department of Neurology and Institute of Neuropathology, RWTH Aachen University, Aachen, Germany; Department of Neurology, University Hospitals Leuven and University of Leuven (KU Leuven), Leuven, Belgium
| | - Rudolf A Kley
- Department of Neurology, Heimer Institute for Muscle Research, Ruhr-University Bochum, Bochum, Germany
| | - Benno Kusters
- Department of Pathology, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Simone Schrading
- Department of Radiology, RWTH Aachen University, Aachen, Germany
| | - Erik J Kamsteeg
- Department of Human Genetics, Radboud University Medical Centre, The Netherlands
| | - Nicol C Voermans
- Department of Neurology, Radboud University Medical Centre, Nijmegen, The Netherlands.
| |
Collapse
|
47
|
Begay RL, Tharp CA, Martin A, Graw SL, Sinagra G, Miani D, Sweet ME, Slavov DB, Stafford N, Zeller MJ, Alnefaie R, Rowland TJ, Brun F, Jones KL, Gowan K, Mestroni L, Garrity DM, Taylor MRG. FLNC Gene Splice Mutations Cause Dilated Cardiomyopathy. JACC Basic Transl Sci 2016; 1:344-359. [PMID: 28008423 PMCID: PMC5166708 DOI: 10.1016/j.jacbts.2016.05.004] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A genetic etiology has been identified in 30% to 40% of dilated cardiomyopathy (DCM) patients, yet only 50% of these cases are associated with a known causative gene variant. Thus, in order to understand the pathophysiology of DCM, it is necessary to identify and characterize additional genes. In this study, whole exome sequencing in combination with segregation analysis was used to identify mutations in a novel gene, filamin C (FLNC), resulting in a cardiac-restricted DCM pathology. Here we provide functional data via zebrafish studies and protein analysis to support a model implicating FLNC haploinsufficiency as a mechanism of DCM. Deoxyribonucleic acid obtained from 2 large DCM families was studied using whole-exome sequencing and cosegregation analysis resulting in the identification of a novel disease gene, FLNC. The 2 families, from the same Italian region, harbored the same FLNC splice-site mutation (FLNC c.7251+1G>A). A third U.S. family was then identified with a novel FLNC splice-site mutation (FLNC c.5669-1delG) that leads to haploinsufficiency as shown by the FLNC Western blot analysis of the heart muscle. The FLNC ortholog flncb morpholino was injected into zebrafish embryos, and when flncb was knocked down caused a cardiac dysfunction phenotype. On electron microscopy, the flncb morpholino knockdown zebrafish heart showed defects within the Z-discs and sarcomere disorganization.
Collapse
Affiliation(s)
- Rene L Begay
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Charles A Tharp
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - August Martin
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, CO
| | - Sharon L Graw
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Gianfranco Sinagra
- Cardiovascular Department, Ospedali Riuniti and University of Trieste, Trieste, Italy
| | - Daniela Miani
- Department of Cardiothoracic Science, University Hospital S. Maria della Misericordia, Udine, Italy
| | - Mary E Sweet
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Dobromir B Slavov
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Neil Stafford
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, CO; Cardiovascular and Biofluid Mechanics Laboratory, Colorado State University, Fort Collins, CO
| | - Molly J Zeller
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, CO
| | - Rasha Alnefaie
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, CO
| | - Teisha J Rowland
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Francesca Brun
- Cardiovascular Department, Ospedali Riuniti and University of Trieste, Trieste, Italy
| | - Kenneth L Jones
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO
| | - Katherine Gowan
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Aurora, CO
| | - Luisa Mestroni
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| | - Deborah M Garrity
- Center for Cardiovascular Research and Department of Biology, Colorado State University, Fort Collins, CO
| | - Matthew R G Taylor
- Cardiovascular Institute and Adult Medical Genetics Program, University of Colorado Denver, Aurora, CO
| |
Collapse
|
48
|
Ruparelia AA, Oorschot V, Ramm G, Bryson-Richardson RJ. FLNC myofibrillar myopathy results from impaired autophagy and protein insufficiency. Hum Mol Genet 2016; 25:2131-2142. [PMID: 26969713 DOI: 10.1093/hmg/ddw080] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Accepted: 03/07/2016] [Indexed: 01/23/2023] Open
Abstract
Myofibrillar myopathy is a progressive muscle disease characterized by the disintegration of muscle fibers and formation of protein aggregates. Causative mutations have been identified in nine genes encoding Z-disk proteins, including the actin binding protein filamin C (FLNC). To investigate the mechanism of disease in FLNCW2710X myopathy we overexpressed fluorescently tagged FLNC or FLNCW2710X in zebrafish. Expression of FLNCW2710X causes formation of protein aggregates but surprisingly, our studies reveal that the mutant protein localizes correctly to the Z-disk and is capable of rescuing the fiber disintegration phenotype that results from FLNC knockdown. This demonstrates that the functions necessary for muscle integrity are not impaired, and suggests that it is the formation of protein aggregates and subsequent sequestration of FLNC away from the Z-disk that results in myofibrillar disintegration. Similar to those found in patients, the aggregates in FLNCW2710X expressing fish contain the co-chaperone BAG3. FLNC is a target of the BAG3-mediated chaperone assisted selective autophagy (CASA) pathway and therefore we investigated its role, and the role of autophagy in general, in clearing protein aggregates. We reveal that despite BAG3 recruitment to the aggregates they are not degraded via CASA. Additionally, recruitment of BAG3 is sufficient to block alternative autophagy pathways which would otherwise clear the aggregates. This blockage can be relieved by reducing BAG3 levels or by stimulating autophagy. This study therefore identifies both BAG3 reduction and autophagy promotion as potential therapies for FLNCW2710X myofibrillar myopathy, and identifies protein insufficiency due to sequestration, compounded by impaired autophagy, as the cause.
Collapse
Affiliation(s)
| | - Viola Oorschot
- The Clive and Vera Ramaciotti Centre for Structural Cryo-Electron Microscopy and and
| | - Georg Ramm
- The Clive and Vera Ramaciotti Centre for Structural Cryo-Electron Microscopy and and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia
| | | |
Collapse
|
49
|
New insights into the protein aggregation pathology in myotilinopathy by combined proteomic and immunolocalization analyses. Acta Neuropathol Commun 2016; 4:8. [PMID: 26842778 PMCID: PMC4739336 DOI: 10.1186/s40478-016-0280-0] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2016] [Accepted: 01/23/2016] [Indexed: 01/09/2023] Open
Abstract
Introduction Myofibrillar myopathies are characterized by progressive muscle weakness and impressive abnormal protein aggregation in muscle fibers. In about 10 % of patients, the disease is caused by mutations in the MYOT gene encoding myotilin. The aim of our study was to decipher the composition of protein deposits in myotilinopathy to get new information about aggregate pathology. Results Skeletal muscle samples from 15 myotilinopathy patients were included in the study. Aggregate and control samples were collected from muscle sections by laser microdissection and subsequently analyzed by a highly sensitive proteomic approach that enables a relative protein quantification. In total 1002 different proteins were detected. Seventy-six proteins showed a significant over-representation in aggregate samples including 66 newly identified aggregate proteins. Z-disc-associated proteins were the most abundant aggregate components, followed by sarcolemmal and extracellular matrix proteins, proteins involved in protein quality control and degradation, and proteins with a function in actin dynamics or cytoskeletal transport. Forty over-represented proteins were evaluated by immunolocalization studies. These analyses validated our mass spectrometric data and revealed different regions of protein accumulation in abnormal muscle fibers. Comparison of data from our proteomic analysis in myotilinopathy with findings in other myofibrillar myopathy subtypes indicates a characteristic basic pattern of aggregate composition and resulted in identification of a highly sensitive and specific diagnostic marker for myotilinopathy. Conclusions Our findings i) indicate that main protein components of aggregates belong to a network of interacting proteins, ii) provide new insights into the complex regulation of protein degradation in myotilinopathy that may be relevant for new treatment strategies, iii) imply a combination of a toxic gain-of-function leading to myotilin-positive protein aggregates and a loss-of-function caused by a shift in subcellular distribution with a deficiency of myotilin at Z-discs that impairs the integrity of myofibrils, and iv) demonstrate that proteomic analysis can be helpful in differential diagnosis of protein aggregate myopathies. Electronic supplementary material The online version of this article (doi:10.1186/s40478-016-0280-0) contains supplementary material, which is available to authorized users.
Collapse
|
50
|
Orfanos Z, Gödderz MPO, Soroka E, Gödderz T, Rumyantseva A, van der Ven PFM, Hawke TJ, Fürst DO. Breaking sarcomeres by in vitro exercise. Sci Rep 2016; 6:19614. [PMID: 26804343 PMCID: PMC4726327 DOI: 10.1038/srep19614] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 12/16/2015] [Indexed: 11/30/2022] Open
Abstract
Eccentric exercise leads to focal disruptions in the myofibrils, referred to as “lesions”. These structures are thought to contribute to the post-exercise muscle weakness, and to represent areas of mechanical damage and/or remodelling. Lesions have been investigated in human biopsies and animal samples after exercise. However, this approach does not examine the mechanisms behind lesion formation, or their behaviour during contraction. To circumvent this, we used electrical pulse stimulation (EPS) to simulate exercise in C2C12 myotubes, combined with live microscopy. EPS application led to the formation of sarcomeric lesions in the myotubes, resembling those seen in exercised mice, increasing in number with the time of application or stimulation intensity. Furthermore, transfection with an EGFP-tagged version of the lesion and Z-disc marker filamin-C allowed us to observe the formation of lesions using live cell imaging. Finally, using the same technique we studied the behaviour of these structures during contraction, and observed them to be passively stretching. This passive behaviour supports the hypothesis that lesions contribute to the post-exercise muscle weakness, protecting against further damage. We conclude that EPS can be reliably used as a model for the induction and study of sarcomeric lesions in myotubes in vitro.
Collapse
Affiliation(s)
- Zacharias Orfanos
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Markus P O Gödderz
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Ekaterina Soroka
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Tobias Gödderz
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Anastasia Rumyantseva
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Peter F M van der Ven
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| | - Thomas J Hawke
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, ON, L8N 3Z5, Canada
| | - Dieter O Fürst
- Institute for Cell Biology, University of Bonn, Ulrich-Haberland-Str. 61a, 53121 Bonn, Germany
| |
Collapse
|