1
|
Haliloğlu G, Ravenscroft G. The evolving genetic landscape of neuromuscular fetal akinesias. J Neuromuscul Dis 2025:22143602251339357. [PMID: 40356365 DOI: 10.1177/22143602251339357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Fetal akinesia is a broad term used to describe absent (or reduced, fetal hypokinesia) fetal movements, and it can be detected as early as the first trimester. Depending on the developmental age of onset, anything that interferes or limits the normal in utero movement results in a range of deformations affecting multiple organs and organ systems. Arthrogryposis, also termed arthrogryposis multiplex congenita (AMC), is a definitive terminology for multiple congenital contractures, with two major subgroups; amyoplasia and distal arthrogryposis (DA). The spectrum includes fetal akinesia deformation sequence (FADS), lethal congenital contracture syndrome (LCCS), and multiple pterygium syndrome (MPS). Variants in more than >400 genes are known to cause AMC, and it is increasingly recognized that variants in genes encoding critical components (including ventral horn cell, peripheral nerve, neuromuscular junction, skeletal muscle) of the extended motor unit underlie ∼40% of presentations. With unbiased screening approaches, including sequencing of comprehensive disease gene panels, exomes and genomes, novel genes and phenotypic expansions associated with known human disease genes have been uncovered in the setting of fetal akinesia. Autosomal-recessive titinopathy is the most frequent genetic cause of AMC. Accurate genetic diagnosis is critical to genetic counseling and informing family planning. Around 50% remain undiagnosed following comprehensive prenatal, diagnostic or research screening. Comprehensive phenotyping and periodic reanalysis with appropriate genomic tools are valuable strategies when faced with initial inconclusive results. There are likely many novel causative genes still to identify, which will inform our understanding of the molecular pathways underlying early human development and in utero movement.
Collapse
Affiliation(s)
- Göknur Haliloğlu
- Division of Pediatric Neurology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Gianina Ravenscroft
- Centre for Medical Research, Faculty of Health and Medical Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Rare Disease Genetics and Functional Genomics, Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia
| |
Collapse
|
2
|
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
|
3
|
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
|
4
|
E.S K, A.K Z, M.Yu S, K.I P, N.L R, E.G N, K.S S, A.A K, T.L V, A.S M, E.N M, T.M P, E.S V, A.A K. Distinct molecular features of FLNC mutations, associated with different clinical phenotypes. Cytoskeleton (Hoboken) 2025; 82:158-174. [PMID: 39315490 PMCID: PMC11904857 DOI: 10.1002/cm.21922] [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: 03/04/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/25/2024]
Abstract
Filamin С is a key an actin-binding protein of muscle cells playing a critical role in maintaining structural integrity and sarcomere organization. FLNC mutations contribute to various types of cardiomyopathies and myopathies through potentially different molecular mechanisms. Here, we described the impact of two clinically distinct FLNC variants (R1267Q associated with arrhythmogenic cardiomyopathy and V2264M associated with restrictive cardiomyopathy) on calcium homeostasis, electrophysiology, and gene expression profile of iPSC-derived patient-specific cardiomyocytes. We demonstrated that R1267Q FLNC variant leads to greater disturbances in calcium dynamics, Nav1.5 kinetics and action potentials compared to V2264M variant. These functional characteristics were accompanied by transcriptome changes in genes linked to action potential and sodium transport as well as structural cardiomyocyte genes. We suggest distinct molecular effects of two FLNC variants linked to different types of cardiomyopathies in terms of myofilament structure, electrophysiology, ion channel function and intracellular calcium homeostasis providing the molecular the bases for their different clinical phenotypes.
Collapse
Affiliation(s)
- Klimenko E.S
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Zaytseva A.K
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Sorokina M.Yu
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Perepelina K.I
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Rodina N.L
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Nikitina E.G
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Sukhareva K.S
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Khudiakov A.A
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Vershinina T.L
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Muravyev A.S
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Mikhaylov E.N
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Pervunina T.M
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Vasichkina E.S
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
| | - Kostareva A.A
- Almazov National Medical Research CentreInstitute of Molecular Biology and GeneticsSaint‐PetersburgRussia
- Karolinska Institutet, Department of Women's and Children's HealthKarolinska University HospitalStockholmSweden
| |
Collapse
|
5
|
Sun W, Yan H, Sun M, Wang J, Li K. Expanding the clinical spectrum of 19p13.3 microduplication syndrome: a case report highlighting nephrotic syndrome and literature review. BMC Pediatr 2025; 25:70. [PMID: 39875952 PMCID: PMC11773902 DOI: 10.1186/s12887-025-05394-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Accepted: 01/03/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Common clinical findings in patients with 19p13.3 duplication include intrauterine growth restriction, intellectual disability, developmental delay, microcephaly, and distinctive facial features. In this study, we report the case of a patient with 19p13.3 microduplication and novel clinical findings, specifically nephrotic syndrome. CASE PRESENTATIONS A 4-year-old girl was admitted to our hospital in December 2020 with a fever and cough that had persisted for 3 days. A series of treatments, chromosomal microarray analysis (CMA) and whole exome sequencing (WES) were performed. Relevant literature was reviewed using the search terms "19p13.3" and "19p13.3 microduplication syndrome" in the China Knowledge Network, Wanfang Database, Weipu Journal Service Platform, and PubMed (date range: database establishment to September 2023). In addition to common symptoms, such as developmental delay, microcephaly, distinctive facial features, and congenital heart defects, the patient also had nephrotic syndrome, a previously unreported phenomenon. CMA results showed a 3.6 Mb fragment duplication (copy number: 3) in the chr19p13.3 region, containing 127 protein-coding genes (including CELF5, NFIC, SMIM24, PIAS4, ATCAY, MAP2K2, and ZBTB7A). WES revealed a filamin C mutation (p.Glu309Valfs × 11). The mutation status of the patient and her father was heterozygous, whereas the mutation was not detected in the mother. CONCLUSION Microduplication in the 19p13.3 region could be one of the genetic factors contributing to the observed clinical phenotypes. However, patients with developmental delay, microcephaly, distinctive facial features, congenital heart defects, and urogenital system disorders may exhibit these manifestations due to various genetic syndromes; therefore, simply considering the possibility of 19p13.3 microduplication syndrome based on these non-specific features is not sufficient. Further comprehensive evaluations, including CMA, should be conducted in conjunction with other genetic tests and detailed clinical examinations to accurately determine the underlying genetic causes.
Collapse
Affiliation(s)
- Wenjie Sun
- Pediatric Internal Medicine, Yantai Yuhuangding Hospital, No.20 Yuhuangding East Road, Zhifu District, Yantai City, Shandong, 264000, China
| | - Hong Yan
- Pediatric Internal Medicine, Yantai Yuhuangding Hospital, No.20 Yuhuangding East Road, Zhifu District, Yantai City, Shandong, 264000, China
| | - Mengxin Sun
- Pediatric Internal Medicine, Yantai Yuhuangding Hospital, No.20 Yuhuangding East Road, Zhifu District, Yantai City, Shandong, 264000, China
| | - Jie Wang
- Pediatric Internal Medicine, Yantai Yuhuangding Hospital, No.20 Yuhuangding East Road, Zhifu District, Yantai City, Shandong, 264000, China
| | - Kunxia Li
- Pediatric Internal Medicine, Yantai Yuhuangding Hospital, No.20 Yuhuangding East Road, Zhifu District, Yantai City, Shandong, 264000, China.
| |
Collapse
|
6
|
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
|
7
|
Azab B, Aburizeg D, Shaaban ST, Ji W, Mustafa L, Isbeih NJ, Al-Akily AS, Mohammad H, Jeffries L, Khokha M, Lakhani SA, Al-Ammouri I. Unraveling the genetic tapestry of pediatric sarcomeric cardiomyopathies and masquerading phenocopies in Jordan. Sci Rep 2024; 14:15141. [PMID: 38956129 PMCID: PMC11219879 DOI: 10.1038/s41598-024-64921-9] [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: 12/11/2023] [Accepted: 06/14/2024] [Indexed: 07/04/2024] Open
Abstract
Pediatric cardiomyopathies are mostly attributed to variants in sarcomere-related genes. Unfortunately, the genetic architecture of pediatric cardiomyopathies has never been previously studied in Jordan. We sought to uncover the genetic landscape of 14 patients from nine families with several subtypes of pediatric cardiomyopathies in Jordan using Exome sequencing (ES). Our investigation identified pathogenic and likely pathogenic variants in seven out of nine families (77.8%), clustering in sarcomere-related genes. Surprisingly, phenocopies of sarcomere-related hypertrophic cardiomyopathies were evident in probands with glycogen storage disorder and mitochondrial-related disease. Our study underscored the significance of streamlining ES or expanding cardiomyopathy-related gene panels to identify plausible phenocopies of sarcomere-related cardiomyopathies. Our findings also pointed out the need for genetic testing in patients with cardiomyopathy and their at-risk family members. This can potentially lead to better management strategies, enabling early interventions, and ultimately enhancing their prognosis. Finally, our findings provide an initial contribution to the currently absent knowledge about the molecular underpinnings of cardiomyopathies in Jordan.
Collapse
Affiliation(s)
- Bilal Azab
- Division of Pathology and Laboratory Medicine, Phoenix Children's Hospital, Phoenix, AZ, 85016, USA.
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan.
| | - Dunia Aburizeg
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Sherin T Shaaban
- Department of Biology and Biotechnology, Faculty of Science, American University of Madaba, Madaba, 11821, Jordan
| | - Weizhen Ji
- Department of Pediatrics, Pediatric Genomics Discovery Program, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Lina Mustafa
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Nooredeen Jamal Isbeih
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Amal Saleh Al-Akily
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Hashim Mohammad
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, 11942, Jordan
| | - Lauren Jeffries
- Department of Pediatrics, Pediatric Genomics Discovery Program, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Mustafa Khokha
- Department of Pediatrics, Pediatric Genomics Discovery Program, Yale University School of Medicine, New Haven, CT, 06510, USA
- Department of Genetics, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Saquib A Lakhani
- Department of Pediatrics, Pediatric Genomics Discovery Program, Yale University School of Medicine, New Haven, CT, 06510, USA
| | - Iyad Al-Ammouri
- Department of Pediatrics, School of Medicine, The University of Jordan, Amman, 11942, Jordan.
| |
Collapse
|
8
|
Malikides O, Simantirakis E, Zacharis E, Fragkiadakis K, Kochiadakis G, Marketou M. Cardiac Remodeling and Ventricular Pacing: From Genes to Mechanics. Genes (Basel) 2024; 15:671. [PMID: 38927607 PMCID: PMC11203142 DOI: 10.3390/genes15060671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 05/17/2024] [Accepted: 05/21/2024] [Indexed: 06/28/2024] Open
Abstract
Cardiac remodeling and ventricular pacing represent intertwined phenomena with profound implications for cardiovascular health and therapeutic interventions. This review explores the intricate relationship between cardiac remodeling and ventricular pacing, spanning from the molecular underpinnings to biomechanical alterations. Beginning with an examination of genetic predispositions and cellular signaling pathways, we delve into the mechanisms driving myocardial structural changes and electrical remodeling in response to pacing stimuli. Insights into the dynamic interplay between pacing strategies and adaptive or maladaptive remodeling processes are synthesized, shedding light on the clinical implications for patients with various cardiovascular pathologies. By bridging the gap between basic science discoveries and clinical translation, this review aims to provide a comprehensive understanding of cardiac remodeling in the context of ventricular pacing, paving the way for future advancements in cardiovascular care.
Collapse
Affiliation(s)
- Onoufrios Malikides
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
| | - Emmanouel Simantirakis
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
- Medical School, University of Crete, 71003 Heraklion, Greece
| | - Evangelos Zacharis
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
- Medical School, University of Crete, 71003 Heraklion, Greece
| | - Konstantinos Fragkiadakis
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
- Medical School, University of Crete, 71003 Heraklion, Greece
| | - George Kochiadakis
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
- Medical School, University of Crete, 71003 Heraklion, Greece
| | - Maria Marketou
- Department of Cardiology, University General Hospital of Heraklion, 71003 Heraklion, Greece; (E.S.); (E.Z.); (K.F.); (G.K.); (M.M.)
- Medical School, University of Crete, 71003 Heraklion, Greece
| |
Collapse
|
9
|
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
|
10
|
Inoue S, Morita H. Characteristic Imaging Phenotype of Arrhythmogenic Cardiomyopathy With Filamin C Gene Variant. Circ J 2023; 87:1404-1405. [PMID: 37673657 DOI: 10.1253/circj.cj-23-0616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Affiliation(s)
- Shunsuke Inoue
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| | - Hiroyuki Morita
- Department of Cardiovascular Medicine, Graduate School of Medicine, The University of Tokyo
| |
Collapse
|
11
|
Kumar P, Paramasivam G, Prabhu MA, Devasia T, Rajasekhar M. A novel FLNC variation associated with restrictive cardiomyopathy with an unusually long clinical course — A case report. GENE REPORTS 2023. [DOI: 10.1016/j.genrep.2023.101769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
12
|
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
|
13
|
Schnabel F, Schuler E, Al-Maawali A, Chaurasia A, Syrbe S, Al-Kindi A, Bhavani GS, Shukla A, Altmüller J, Nürnberg P, Banka S, Girisha KM, Li Y, Wollnik B, Yigit G. Homozygous loss-of-function variants in FILIP1 cause autosomal recessive arthrogryposis multiplex congenita with microcephaly. Hum Genet 2023; 142:543-552. [PMID: 36943452 PMCID: PMC10060356 DOI: 10.1007/s00439-023-02528-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 01/21/2023] [Indexed: 03/23/2023]
Abstract
Arthrogryposis multiplex congenita forms a broad group of clinically and etiologically heterogeneous disorders characterized by congenital joint contractures that involve at least two different parts of the body. Neurological and muscular disorders are commonly underlying arthrogryposis. Here, we report five affected individuals from three independent families sharing an overlapping phenotype with congenital contractures affecting shoulder, elbow, hand, hip, knee and foot as well as scoliosis, reduced palmar and plantar skin folds, microcephaly and facial dysmorphism. Using exome sequencing, we identified homozygous truncating variants in FILIP1 in all patients. FILIP1 is a regulator of filamin homeostasis required for the initiation of cortical cell migration in the developing neocortex and essential for the differentiation process of cross-striated muscle cells during myogenesis. In summary, our data indicate that bi-allelic truncating variants in FILIP1 are causative of a novel autosomal recessive disorder and expand the spectrum of genetic factors causative of arthrogryposis multiplex congenita.
Collapse
Affiliation(s)
- Franziska Schnabel
- Institute of Human Genetics, University Medical Center Göttingen, Heinrich-Düker-Weg 12, 37073, Göttingen, Germany
- Institute of Human Genetics, University of Leipzig Hospitals and Clinics, 04103, Leipzig, Germany
| | - Elisabeth Schuler
- Division of Paediatric Epileptology, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Almundher Al-Maawali
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
- Genetic and Developmental Medicine Clinic, Sultan Qaboos University Hospital, Muscat, Oman
| | - Ankur Chaurasia
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
| | - Steffen Syrbe
- Division of Paediatric Epileptology, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120, Heidelberg, Germany
| | - Adila Al-Kindi
- Department of Genetics, College of Medicine and Health Sciences, Sultan Qaboos University, Muscat, Oman
- Genetic and Developmental Medicine Clinic, Sultan Qaboos University Hospital, Muscat, Oman
| | - Gandham SriLakshmi Bhavani
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Anju Shukla
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Janine Altmüller
- Cologne Center for Genomics (CCG), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Core Facility Genomics, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Peter Nürnberg
- Cologne Center for Genomics (CCG), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Siddharth Banka
- Division of Evolution, Infection and Genomics, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PL, UK
- Manchester Centre for Genomic Medicine, Health Innovation Manchester, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, M13 9WL, UK
| | - Katta M Girisha
- Department of Medical Genetics, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal, India
| | - Yun Li
- Institute of Human Genetics, University Medical Center Göttingen, Heinrich-Düker-Weg 12, 37073, Göttingen, Germany
| | - Bernd Wollnik
- Institute of Human Genetics, University Medical Center Göttingen, Heinrich-Düker-Weg 12, 37073, Göttingen, Germany.
- Cluster of Excellence "Multiscale Bioimaging: From Molecular Machines To Networks of Excitable Cells" (MBExC), University of Göttingen, 37073, Göttingen, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
| | - Gökhan Yigit
- Institute of Human Genetics, University Medical Center Göttingen, Heinrich-Düker-Weg 12, 37073, Göttingen, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany.
| |
Collapse
|
14
|
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
|
15
|
A Missense Variant in PDK1 Associated with Severe Neurodevelopmental Delay and Epilepsy. Biomedicines 2022; 10:biomedicines10123171. [PMID: 36551928 PMCID: PMC9775741 DOI: 10.3390/biomedicines10123171] [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: 09/30/2022] [Revised: 12/02/2022] [Accepted: 12/04/2022] [Indexed: 12/13/2022] Open
Abstract
The pyruvate dehydrogenase complex (PDC) is responsible for the conversion of pyruvate into acetyl-CoA, which is used for energy conversion in cells. PDC activity is regulated by phosphorylation via kinases and phosphatases (PDK/PDP). Variants in all subunits of the PDC and in PDK3 have been reported, with varying phenotypes including lactic acidosis, neurodevelopmental delay, peripheral neuropathy, or seizures. Here, we report a de novo heterozygous missense variant in PDK1 (c.1139G > A; p.G380D) in a girl with developmental delay and early onset severe epilepsy. To investigate the role of PDK1G380D in energy metabolism and neuronal development, we used a zebrafish model. In zebrafish embryos we show a reduced number of cells with mitochondria with membrane potential, reduced movements, and a delay in neuronal development. Furthermore, we observe a reduction in the phosphorylation of PDH-E1α by PDKG380D, which suggests a disruption in the regulation of PDC activity. Finally, in patient fibroblasts, a mild reduction in the ratio of phosphorylated PDH over total PDH-E1α was detected. In summary, our findings support the notion that this aberrant PDK1 activity is the cause of clinical symptoms in the patient.
Collapse
|
16
|
Cardiovascular Involvement in Pediatric FLNC Variants: A Case Series of Fourteen Patients. J Cardiovasc Dev Dis 2022; 9:jcdd9100332. [PMID: 36286284 PMCID: PMC9604120 DOI: 10.3390/jcdd9100332] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 09/22/2022] [Indexed: 11/16/2022] Open
Abstract
Filamin C is a protein specifically expressed in myocytes and cardiomyocytes and is involved in several biological functions, including sarcomere contractile activity, signaling, cellular adhesion, and repair. FLNC variants are associated with different disorders ranging from striated muscle (myofibrillar distal or proximal) myopathy to cardiomyopathies (CMPs) (restrictive, hypertrophic, and dilated), or both. The outcome depends on functional consequences of the detected variants, which result either in FLNC haploinsufficiency or in an aberrant protein, the latter affecting sarcomere structure leading to protein aggregates. Cardiac manifestations of filaminopathies are most often described as adult onset CMPs and limited reports are available in children or on other cardiac spectrums (congenital heart defects-CHDs, or arrhythmias). Here we report on 13 variants in 14 children (2.8%) out of 500 pediatric patients with early-onset different cardiac features ranging from CMP to arrhythmias and CHDs. In one patient, we identified a deletion encompassing FLNC detected by microarray, which was overlooked by next generation sequencing. We established a potential genotype-phenotype correlation of the p.Ala1186Val variant in severe and early-onset restrictive cardiomyopathy (RCM) associated with a limb-girdle defect (two new patients in addition to the five reported in the literature). Moreover, in three patients (21%), we identified a relatively frequent finding of long QT syndrome (LQTS) associated with RCM (n = 2) and a hypertrabeculated left ventricle (n = 1). RCM and LQTS in children might represent a specific red flag for FLNC variants. Further studies are warranted in pediatric cohorts to delineate potential expanding phenotypes related to FLNC.
Collapse
|
17
|
Muravyev A, Vershinina T, Tesner P, Sjoberg G, Fomicheva Y, Čajbiková NN, Kozyreva A, Zhuk S, Mamaeva E, Tarnovskaya S, Jornholt J, Sokolnikova P, Pervunina T, Vasichkina E, Sejersen T, Kostareva A. Rare clinical phenotype of filaminopathy presenting as restrictive cardiomyopathy and myopathy in childhood. Orphanet J Rare Dis 2022; 17:358. [PMID: 36104822 PMCID: PMC9476594 DOI: 10.1186/s13023-022-02477-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/13/2022] [Indexed: 11/10/2022] Open
Abstract
Background FLNC is one of the few genes associated with all types of cardiomyopathies, but it also underlies neuromuscular phenotype. The combination of concomitant neuromuscular and cardiac involvement is not often observed in filaminopathies and the impact of this on the disease prognosis has hitherto not been analyzed. Results Here we provide a detailed clinical, genetic, and structural prediction analysis of distinct FLNC-associated phenotypes based on twelve pediatric cases. They include early-onset restrictive cardiomyopathy (RCM) in association with congenital myopathy. In all patients the initial diagnosis was established during the first year of life and in five out of twelve (41.7%) patients the first symptoms were observed at birth. RCM was present in all patients, often in combination with septal defects. No ventricular arrhythmias were noted in any of the patients presented here. Myopathy was confirmed by neurological examination, electromyography, and morphological studies. Arthrogryposes was diagnosed in six patients and remained clinically meaningful with increasing age in three of them. One patient underwent successful heart transplantation at the age of 18 years and two patients are currently included in the waiting list for heart transplantation. Two died due to congestive heart failure. One patient had ICD instally as primary prevention of SCD. In ten out of twelve patients the disease was associated with missense variants and only in two cases loss of function variants were detected. In half of the described cases, an amino acid substitution A1186V, altering the structure of IgFLNc10, was found. Conclusions The present description of twelve cases of early-onset restrictive cardiomyopathy with congenital myopathy and FLNC mutation, underlines a distinct unique phenotype that can be suggested as a separate clinical form of filaminopathies. Amino acid substitution A1186V, which was observed in half of the cases, defines a mutational hotspot for the reported combination of myopathy and cardiomyopathy. Several independent molecular mechanisms of FLNC mutations linked to filamin structure and function can explain the broad spectrum of FLNC-associated phenotypes. Early disease presentation and unfavorable prognosis of heart failure demanding heart transplantation make awareness of this clinical form of filaminopathy of great clinical importance. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02477-5.
Collapse
|
18
|
Clinical Exome Sequencing Revealed a De Novo FLNC Mutation in a Child with Restrictive Cardiomyopathy. CARDIOGENETICS 2022. [DOI: 10.3390/cardiogenetics12020019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Restrictive cardiomyopathy (RCM) is a rare disease of the myocardium caused by mutations in several genes including TNNT2, DES, TNNI3, MYPN and FLNC. Individuals affected by RCM often develop heart failure at a young age, requiring early heart transplantation. A 7-year-old patient was referred for genetic testing following a diagnosis of restrictive cardiomyopathy. Clinical exome sequencing analysis identified a likely pathogenic mutation in the FLNC gene [(NM_001458.5 c.6527_6547dup p.(Arg2176_2182dup)]. Its clinical relevance was augmented by the fact that this variant was absent in the parents and was thus interpreted as de novo. Genetic testing is a powerful tool to clarify the diagnosis, guide intervention strategies and enable cascade testing in patients with pediatric-onset RCM.
Collapse
|
19
|
Zheng M, Huang H, Zhu X, Ho H, Li L, Ji X. Clinical genetic testing in four highly suspected pediatric restrictive cardiomyopathy cases. BMC Cardiovasc Disord 2022; 22:240. [PMID: 35614389 PMCID: PMC9131548 DOI: 10.1186/s12872-022-02675-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 05/13/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Restrictive cardiomyopathy (RCM) presents a high risk for sudden cardiac death in pediatric patients. Constrictive pericarditis (CP) exhibits a similar clinical presentation to RCM and requires differential diagnosis. While mutations of genes that encode sarcomeric and cytoskeletal proteins may lead to RCM, infection, rather than gene mutation, is the main cause of CP. Genetic testing may be helpful in the clinical diagnosis of RCM. METHODS In this case series study, we screened for TNNI3, TNNT2, and DES gene mutations that are known to be etiologically linked to RCM in four pediatric patients with suspected RCM. RESULTS We identified one novel heterozygous mutation, c.517C>T (substitution, position 517 C → T) (amino acid conversion, p.Leu173Phe), and two already known heterozygous mutations, c.508C>T (substitution, position 508, C → T) (amino acid conversion, p.Arg170Trp) and c.575G>A (substitution, position 575, G → A) (amino acid conversion, p.Arg192His), in the TNNI3 gene in three of the four patients. CONCLUSION Our findings support the notion that genetic testing may be helpful in the clinical diagnosis of RCM.
Collapse
Affiliation(s)
- Min Zheng
- Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Infection and Immunity, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China
| | - Hong Huang
- Pediatric Department, North-Kuanren General Hospital of Chongqing, Chongqing, 401121, China
| | - Xu Zhu
- Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Infection and Immunity, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China
| | - Harvey Ho
- Auckland Bioengineering Institute, The University of Auckland, Private Bag 92019, Auckland, 1142, New Zealand
| | - Liling Li
- Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Infection and Immunity, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China
| | - Xiaojuan Ji
- Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Chongqing Key Laboratory of Child Infection and Immunity, 136 Zhongshan 2nd Road, Yu Zhong District, Chongqing, 400014, China.
| |
Collapse
|
20
|
Genetic Insights into Primary Restrictive Cardiomyopathy. J Clin Med 2022; 11:jcm11082094. [PMID: 35456187 PMCID: PMC9027761 DOI: 10.3390/jcm11082094] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/05/2022] [Accepted: 04/06/2022] [Indexed: 12/04/2022] Open
Abstract
Restrictive cardiomyopathy is a rare cardiac disease causing severe diastolic dysfunction, ventricular stiffness and dilated atria. In consequence, it induces heart failure often with preserved ejection fraction and is associated with a high mortality. Since it is a poor clinical prognosis, patients with restrictive cardiomyopathy frequently require heart transplantation. Genetic as well as non-genetic factors contribute to restrictive cardiomyopathy and a significant portion of cases are of unknown etiology. However, the genetic forms of restrictive cardiomyopathy and the involved molecular pathomechanisms are only partially understood. In this review, we summarize the current knowledge about primary genetic restrictive cardiomyopathy and describe its genetic landscape, which might be of interest for geneticists as well as for cardiologists.
Collapse
|
21
|
Chintanaphol M, Orgil BO, Alberson NR, Towbin JA, Purevjav E. Restrictive cardiomyopathy: from genetics and clinical overview to animal modeling. Rev Cardiovasc Med 2022; 23:108. [PMID: 35345275 DOI: 10.31083/j.rcm2303108] [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: 08/18/2021] [Revised: 10/08/2021] [Accepted: 10/25/2021] [Indexed: 11/06/2022] Open
Abstract
Restrictive cardiomyopathy (RCM), a potentially devastating heart muscle disorder, is characterized by diastolic dysfunction due to abnormal muscle relaxation and myocardial stiffness resulting in restrictive filling of the ventricles. Diastolic dysfunction is often accompanied by left atrial or bi-atrial enlargement and normal ventricular size and systolic function. RCM is the rarest form of cardiomyopathy, accounting for 2-5% of pediatric cardiomyopathy cases, however, survival rates have been reported to be 82%, 80%, and 68% at 1-, 2-, and 5-years after diagnosis, respectively. RCM can be idiopathic, familial, or secondary to a systemic disorder, such as amyloidosis, sarcoidosis, and hereditary hemochromatosis. Approximately 30% of cases are familial RCM, and the genes that have been linked to RCM are cTnT, cTnI, MyBP-C, MYH7, MYL2, MYL3, DES, MYPN, TTN, BAG3, DCBLD2, LNMA, and FLNC. Increased Ca2+ sensitivity, sarcomere disruption, and protein aggregates are some of the few mechanisms of pathogenesis that have been revealed by studies utilizing cell lines and animal models. Additional exploration into the pathogenesis of RCM is necessary to create novel therapeutic strategies to reverse restrictive cardiomyopathic phenotypes.
Collapse
Affiliation(s)
- Michelle Chintanaphol
- College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38103, USA
| | - Buyan-Ochir Orgil
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| | - Neely R Alberson
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| | - Jeffrey A Towbin
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
- Pediatric Cardiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Enkhsaikhan Purevjav
- Heart Institute, Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA
- Children's Foundation Research Institute, Le Bonheur Children's Hospital, Memphis, TN 38103, USA
| |
Collapse
|
22
|
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
|
23
|
Zhang Y, Yan H, Liu J, Yan H, Ma Y, Wei C, Wang Z, Xiong H, Chang X. Clinical and genetic features of infancy-onset congenital myopathies from a Chinese paediatric centre. BMC Pediatr 2022; 22:65. [PMID: 35081925 PMCID: PMC8790871 DOI: 10.1186/s12887-021-03024-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 11/18/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Congenital myopathies are a group of rare neuromuscular diseases characterized by specific histopathological features. The relationship between the pathologies and the genetic causes is complex, and the prevalence of myopathy-causing genes varies among patients from different ethnic groups. The aim of the present study was to characterize congenital myopathies with infancy onset among patients registered at our institution. METHOD This retrospective study enrolled 56 patients based on the pathological and/or genetic diagnosis. Clinical, histopathological and genetic features of the patients were analysed with long-term follow-up. RESULTS Twenty-six out of 43 patients who received next-generation sequencing had genetic confirmation, and RYR1 variations (12/26) were the most prevalent. Eighteen novel variations were identified in 6 disease-causing genes, including RYR1, NEB, TTN, TNNT1, DNM2 and ACTA1. Nemaline myopathy (17/55) was the most common histopathology. The onset ages ranged from birth to 1 year. Thirty-one patients were followed for 3.83 ± 3.05 years (ranging from 3 months to 11 years). No patient died before 1 year. Two patients died at 5 years and 8 years respectively. The motor abilities were stable or improved in 23 patients and deteriorated in 6 patients. Ten (10/31) patients developed respiratory involvement, and 9 patients (9/31) had mildly abnormal electrocardiograms and/or echocardiograms. CONCLUSION The severity of congenital myopathies in the neonatal/infantile period may vary in patients from different ethnic groups. More concern should be given to cardiac monitoring in patients with congenital myopathies even in those with static courses.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Paediatrics, Peking University First Hospital, No.1 Xianmen Street, Xicheng District, 100034, Beijing, PR China.,Department of Paediatrics, Peking University International Hospital, 102206, Beijing, PR China
| | - Hui Yan
- Department of Paediatrics, Peking University First Hospital, No.1 Xianmen Street, Xicheng District, 100034, Beijing, PR China
| | - Jieyu Liu
- Department of Paediatrics, Peking University First Hospital, No.1 Xianmen Street, Xicheng District, 100034, Beijing, PR China
| | - Huifang Yan
- Department of Paediatrics, Peking University First Hospital, No.1 Xianmen Street, Xicheng District, 100034, Beijing, PR China
| | - Yinan Ma
- Department of Central Laboratory, Peking University First Hospital, 100034, Beijing, PR China
| | - Cuijie Wei
- Department of Paediatrics, Peking University First Hospital, No.1 Xianmen Street, Xicheng District, 100034, Beijing, PR China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, 100034, Beijing, PR China
| | - Hui Xiong
- Department of Paediatrics, Peking University First Hospital, No.1 Xianmen Street, Xicheng District, 100034, Beijing, PR China
| | - Xingzhi Chang
- Department of Paediatrics, Peking University First Hospital, No.1 Xianmen Street, Xicheng District, 100034, Beijing, PR China.
| |
Collapse
|
24
|
Brodehl A, Hain C, Flottmann F, Ratnavadivel S, Gaertner A, Klauke B, Kalinowski J, Körperich H, Gummert J, Paluszkiewicz L, Deutsch MA, Milting H. The Desmin Mutation DES-c.735G>C Causes Severe Restrictive Cardiomyopathy by Inducing In-Frame Skipping of Exon-3. Biomedicines 2021; 9:biomedicines9101400. [PMID: 34680517 PMCID: PMC8533191 DOI: 10.3390/biomedicines9101400] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/01/2021] [Accepted: 10/02/2021] [Indexed: 02/02/2023] Open
Abstract
Currently, little is known about the genetic background of restrictive cardiomyopathy (RCM). Herein, we screened an index patient with RCM in combination with atrial fibrillation using a next generation sequencing (NGS) approach and identified the heterozygous mutation DES-c.735G>C. As DES-c.735G>C affects the last base pair of exon-3, it is unknown whether putative missense or splice site mutations are caused. Therefore, we applied nanopore amplicon sequencing revealing the expression of a transcript without exon-3 in the explanted myocardial tissue of the index patient. Western blot analysis verified this finding at the protein level. In addition, we performed cell culture experiments revealing an abnormal cytoplasmic aggregation of the truncated desmin form (p.D214-E245del) but not of the missense variant (p.E245D). In conclusion, we show that DES-c.735G>C causes a splicing defect leading to exon-3 skipping of the DES gene. DES-c.735G>C can be classified as a pathogenic mutation associated with RCM and atrial fibrillation. In the future, this finding might have relevance for the genetic understanding of similar cases.
Collapse
Affiliation(s)
- Andreas Brodehl
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
- Correspondence: (A.B.); (H.M.); Tel.: +49-(0)5731-973530 (A.B.); +49-(0)5731-973510 (H.M.)
| | - Carsten Hain
- Microbial Genomics and Biotechnology, Center for Biotechnology, Bielefeld University, D-33615 Bielefeld, Germany; (C.H.); (J.K.)
| | - Franziska Flottmann
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
| | - Sandra Ratnavadivel
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
| | - Anna Gaertner
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
| | - Bärbel Klauke
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
- Clinic for General and Interventional Cardiology/Angiology, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany
| | - Jörn Kalinowski
- Microbial Genomics and Biotechnology, Center for Biotechnology, Bielefeld University, D-33615 Bielefeld, Germany; (C.H.); (J.K.)
| | - Hermann Körperich
- Heart and Diabetes Center NRW, Institute for Radiology, Nuclear Medicine and Molecular Imaging, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany;
| | - Jan Gummert
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
- Heart and Diabetes Center NRW, Department of Thoracic and Cardiovascular Surgery, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (L.P.); (M.-A.D.)
| | - Lech Paluszkiewicz
- Heart and Diabetes Center NRW, Department of Thoracic and Cardiovascular Surgery, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (L.P.); (M.-A.D.)
| | - Marcus-André Deutsch
- Heart and Diabetes Center NRW, Department of Thoracic and Cardiovascular Surgery, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (L.P.); (M.-A.D.)
| | - Hendrik Milting
- Heart and Diabetes Center NRW, Erich and Hanna Klessmann Institute, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany; (F.F.); (S.R.); (A.G.); (B.K.); (J.G.)
- Correspondence: (A.B.); (H.M.); Tel.: +49-(0)5731-973530 (A.B.); +49-(0)5731-973510 (H.M.)
| |
Collapse
|
25
|
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
|
26
|
Juntas Morales R, Perrin A, Solé G, Lacourt D, Pegeot H, Walther-Louvier U, Cintas P, Cances C, Espil C, Theze C, Zenagui R, Yauy K, Cosset E, Renard D, Rigau V, Maues de Paula A, Uro-Coste E, Arne-Bes MC, Martin Négrier ML, Leboucq N, Acket B, Malfatti E, Biancalana V, Metay C, Richard P, Rendu J, Rivier F, Koenig M, Cossée M. An Integrated Clinical-Biological Approach to Identify Interindividual Variability and Atypical Phenotype-Genotype Correlations in Myopathies: Experience on A Cohort of 156 Families. Genes (Basel) 2021; 12:genes12081199. [PMID: 34440373 PMCID: PMC8392536 DOI: 10.3390/genes12081199] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 07/30/2021] [Indexed: 01/17/2023] Open
Abstract
Diagnosis of myopathies is challenged by the high genetic heterogeneity and clinical overlap of the various etiologies. We previously reported a Next-Generation Sequencing strategy to identify genetic etiology in patients with undiagnosed Limb-Girdle Muscular Dystrophies, Congenital Myopathies, Congenital Muscular Dystrophies, Distal Myopathies, Myofibrillar Myopathies, and hyperCKemia or effort intolerance, using a large gene panel including genes classically associated with other entry diagnostic categories. In this study, we report the comprehensive clinical-biological strategy used to interpret NGS data in a cohort of 156 pediatric and adult patients, that included Copy Number Variants search, variants filtering and interpretation according to ACMG guidelines, segregation studies, deep phenotyping of patients and relatives, transcripts and protein studies, and multidisciplinary meetings. Genetic etiology was identified in 74 patients, a diagnostic yield (47.4%) similar to previous studies. We identified 18 patients (10%) with causative variants in different genes (ACTA1, RYR1, NEB, TTN, TRIP4, CACNA1S, FLNC, TNNT1, and PAPBN1) that resulted in milder and/or atypical phenotypes, with high intrafamilial variability in some cases. Mild phenotypes could mostly be explained by a less deleterious effect of variants on the protein. Detection of inter-individual variability and atypical phenotype-genotype associations is essential for precision medicine, patient care, and to progress in the understanding of the molecular mechanisms of myopathies.
Collapse
Affiliation(s)
- Raul Juntas Morales
- Explorations Neurologiques et Centre SLA, Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), Centre Hospitalier Universitaire de Montpellier, 34295 Montpellier, France;
- Équipe Accueil EA7402, Institut Universitaire de Recherche Clinique (IURC), Université de Montpellier, 34093 Montpellier, France;
| | - Aurélien Perrin
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34093 Montpellier, France
| | - Guilhem Solé
- Service de Neurologie, Centre Hospitalier Universitaire de Bordeaux, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 33000 Bordeaux, France;
| | - Delphine Lacourt
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
| | - Henri Pegeot
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
| | - Ulrike Walther-Louvier
- Service de Neuropédiatrie, Centre Hospitalier Universitaire de Montpellier, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 34295 Montpellier, France; (U.W.-L.); (F.R.)
| | - Pascal Cintas
- Service de Neurologie, Centre Hospitalier Universitaire de Toulouse, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 31059 Toulouse, France; (P.C.); (M.-C.A.-B.); (B.A.)
| | - Claude Cances
- Service de Neuropédiatrie, Centre Hospitalier Universitaire de Toulouse, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 31059 Toulouse, France;
| | - Caroline Espil
- Service de Neuropédiatrie, Centre Hospitalier de Bordeaux, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 33000 Bordeaux, France;
| | - Corinne Theze
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
| | - Reda Zenagui
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
| | - Kevin Yauy
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
| | - Elodie Cosset
- Équipe Accueil EA7402, Institut Universitaire de Recherche Clinique (IURC), Université de Montpellier, 34093 Montpellier, France;
| | - Dimitri Renard
- Service de Neurologie, Centre Hospitalier Universitaire de Nîmes, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 30029 Nîmes, France;
| | - Valerie Rigau
- Service de Pathologie, Centre Hospitalier Universitaire de Montpellier, Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 34295 Montpellier, France;
| | - Andre Maues de Paula
- Service de Pathologie, Centre Hospitalier Universitaire de Marseille, Centre de Référence des Maladies Neuromusculaires PACA-Réunion-Rhône Alpes, 13005 Marseille, France;
| | - Emmanuelle Uro-Coste
- Service de Pathologie, Centre Hospitalier Universitaire de Toulouse, Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 31300 Toulouse, France;
| | - Marie-Christine Arne-Bes
- Service de Neurologie, Centre Hospitalier Universitaire de Toulouse, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 31059 Toulouse, France; (P.C.); (M.-C.A.-B.); (B.A.)
| | - Marie-Laure Martin Négrier
- CHU de Bordeaux, Institut des Maladies Neurodégénératives, Université de Bordeaux, UMR 5293, 33076 Bordeaux, France;
| | - Nicolas Leboucq
- Service de Neuroradiologie, Centre Hospitalier de Montpellier, Centre de Référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 34295 Montpellier, France;
| | - Blandine Acket
- Service de Neurologie, Centre Hospitalier Universitaire de Toulouse, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 31059 Toulouse, France; (P.C.); (M.-C.A.-B.); (B.A.)
| | - Edoardo Malfatti
- Service Neurologie Médicale, Centre de Référence Maladies Neuromusculaires Nord-Est-Ile-de-France, CHU Raymond-Poincaré, 92380 Garches, France;
- U1179 UVSQ-INSERM Handicap Neuromusculaire: Physiologie, Biothérapie et Pharmacologie Appliquées, UFR des Sciences de la Santé Simone Veil, Université Versailles-Saint-Quentin-en-Yvelines, 78180 Versailles, France
| | - Valérie Biancalana
- Laboratoire de Diagnostic Génétique, Université de Strasbourg, 67084 Strasbourg, France;
- Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Inserm U1258, CNRS UMR7104, Université de Strasbourg, 67404 Illkirch, France
| | - Corinne Metay
- Unité Fonctionnelle de Cardiogénétique et Myogénétique, Centre de Génétique, Hôpitaux Universitaire Pitié Salpêtrière–Charles Foix, 75651 Paris, France; (C.M.); (P.R.)
| | - Pascale Richard
- Unité Fonctionnelle de Cardiogénétique et Myogénétique, Centre de Génétique, Hôpitaux Universitaire Pitié Salpêtrière–Charles Foix, 75651 Paris, France; (C.M.); (P.R.)
| | - John Rendu
- CHU Grenoble, Université de Grenoble Alpes, Inserm, U1216, GIN, 38706 Saint-Martin-d’Hères, France;
- Unité Médicale de Génétique Moléculaire, Centre Hospitalier, Universitaire Grenoble Alpes, 38043 Saint-Martin-d’Hères, France
| | - François Rivier
- Service de Neuropédiatrie, Centre Hospitalier Universitaire de Montpellier, Centre de référence des Maladies Neuromusculaires AOC (Atlantique-Occitanie-Caraïbe), 34295 Montpellier, France; (U.W.-L.); (F.R.)
| | - Michel Koenig
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34093 Montpellier, France
| | - Mireille Cossée
- Laboratoire de Génétique Moléculaire, Centre Hospitalier Universitaire de Montpellier, 34093 Montpellier, France; (A.P.); (D.L.); (H.P.); (C.T.); (R.Z.); (K.Y.); (M.K.)
- PhyMedExp, Université de Montpellier, INSERM, CNRS, 34093 Montpellier, France
- Correspondence:
| |
Collapse
|
27
|
Khudiakov AA, Panshin DD, Fomicheva YV, Knyazeva AA, Simonova KA, Lebedev DS, Mikhaylov EN, Kostareva AA. Different Expressions of Pericardial Fluid MicroRNAs in Patients With Arrhythmogenic Right Ventricular Cardiomyopathy and Ischemic Heart Disease Undergoing Ventricular Tachycardia Ablation. Front Cardiovasc Med 2021; 8:647812. [PMID: 33816578 PMCID: PMC8017144 DOI: 10.3389/fcvm.2021.647812] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 02/15/2021] [Indexed: 01/15/2023] Open
Abstract
Introduction: Pericardial fluid is enriched with biologically active molecules of cardiovascular origin including microRNAs. Investigation of the disease-specific extracellular microRNAs could shed light on the molecular processes underlying disease development. Arrhythmogenic right ventricular cardiomyopathy (ARVC) is an inherited heart disease characterized by life-threatening arrhythmias and progressive heart failure development. The current data about the association between microRNAs and ARVC development are limited. Methods and Results: We performed small RNA sequence analysis of microRNAs of pericardial fluid samples obtained during transcutaneous epicardial access for ventricular tachycardia (VT) ablation of six patients with definite ARVC and three post-infarction VT patients. Disease-associated microRNAs of pericardial fluid were identified. Five microRNAs (hsa-miR-1-3p, hsa-miR-21-5p, hsa-miR-122-5p, hsa-miR-206, and hsa-miR-3679-5p) were found to be differentially expressed between patients with ARVC and patients with post-infarction VT. Enrichment analysis of differentially expressed microRNAs revealed their close linkage to cardiac diseases. Conclusion: Our data extend the knowledge of pericardial fluid microRNA composition and highlight five pericardial fluid microRNAs potentially linked to ARVC pathogenesis. Further studies are required to confirm the use of pericardial fluid RNA sequencing in differential diagnosis of ARVC.
Collapse
Affiliation(s)
- Aleksandr A Khudiakov
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Daniil D Panshin
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Yulia V Fomicheva
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Anastasia A Knyazeva
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Ksenia A Simonova
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Dmitry S Lebedev
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Bioengineering Systems, Saint Petersburg Electrotechnical University "LETI", Saint Petersburg, Russia
| | - Evgeny N Mikhaylov
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Bioengineering Systems, Saint Petersburg Electrotechnical University "LETI", Saint Petersburg, Russia
| | - Anna A Kostareva
- Institute of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Women's and Children's Health, Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| |
Collapse
|
28
|
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
|
29
|
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
|
30
|
Genetic Restrictive Cardiomyopathy: Causes and Consequences-An Integrative Approach. Int J Mol Sci 2021; 22:ijms22020558. [PMID: 33429969 PMCID: PMC7827163 DOI: 10.3390/ijms22020558] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 01/05/2021] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
The sarcomere as the smallest contractile unit is prone to alterations in its functional, structural and associated proteins. Sarcomeric dysfunction leads to heart failure or cardiomyopathies like hypertrophic (HCM) or restrictive cardiomyopathy (RCM) etc. Genetic based RCM, a very rare but severe disease with a high mortality rate, might be induced by mutations in genes of non-sarcomeric, sarcomeric and sarcomere associated proteins. In this review, we discuss the functional effects in correlation to the phenotype and present an integrated model for the development of genetic RCM.
Collapse
|
31
|
A 300-kb microduplication of 7q36.3 in a patient with triphalangeal thumb-polysyndactyly syndrome combined with congenital heart disease and optic disc coloboma: a case report. BMC Med Genomics 2020; 13:175. [PMID: 33218365 PMCID: PMC7678048 DOI: 10.1186/s12920-020-00821-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 11/09/2020] [Indexed: 12/04/2022] Open
Abstract
Background Triphalangeal thumb-polysyndactyly syndrome (TPT-PS) is a rare well-defined autosomal dominant disorder characterized by long thumbs with three phalanges combined with pre- and postaxial polydactyly/syndactyly of limbs.
By now, the syndrome has been reported in several large families from different ethnic backgrounds, with a high degree of inter- and intrafamilial variability. The genome locus responsible for TPT-PS has been mapped to the 7q36.3 region harboring a long-range sonic hedgehog (SHH) regulatory sequence (ZRS). Both single-nucleotide variants and complete duplications of ZRS were shown to cause TPT-PS and similar limb phenotypes. TPT-PS usually forms as isolated limb pathology not associated with additional malformations, in particular, with cardiovascular abnormalities. Case presentation Here we report on a rare Russian neonatal case of TPT-PS combined with severe congenital heart disease, namely double outlet right ventricle, and microphthalmia with optic disc coloboma. Pedigree analysis revealed TPT-PS of various expressivity in 10 family members throughout five generations, while the cardiac defect and the eye pathology were detected only in the proband. To extend the knowledge on genotype–phenotype spectrum of TPT-PS, the careful clinical and genomic analysis of the family was performed. High-resolution array-based comparative genomic hybridization (array-CGH) revealed a ~ 300 kb microduplication of 7q36.3 locus (arr[GRCh37] 7q36.3(156385810_156684811) × 3) that co-segregated with TPT-PS in the proband and her mother. The duplication encompassed three genes including LMBR1, the intron 5 of which is known to harbor ZRS. Based on whole-exome sequencing data, no additional pathogenic mutations or variants of uncertain clinical significance were found in morbid cardiac genes or genes associated with a microphthalmia/anophthalmia/coloboma spectrum of ocular malformations. Conclusions The results support the previous data, indicating that complete ZRS duplication underlies TPT-PS, and suggest a broader phenotypic impact of the 7q36.3 microduplication. Potential involvement of the 7q36.3 microduplication in the patient’s cardiac and eye malformations is discussed. However, the contribution of some additional genetic/epigenetic factors to the complex patient`s phenotype cannot be excluded entirely. Further comprehensive functional studies are needed to prove the possible involvement of the 7q36.3 locus in congenital heart disease and eye pathology.
Collapse
|
32
|
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
|
33
|
Jorholt J, Formicheva Y, Vershinina T, Kiselev A, Muravyev A, Demchenko E, Fedotov P, Zlotina A, Rygkov A, Vasichkina E, Sejersen T, Kostareva A. Two New Cases of Hypertrophic Cardiomyopathy and Skeletal Muscle Features Associated with ALPK3 Homozygous and Compound Heterozygous Variants. Genes (Basel) 2020; 11:genes11101201. [PMID: 33076350 PMCID: PMC7602582 DOI: 10.3390/genes11101201] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/13/2020] [Accepted: 10/13/2020] [Indexed: 12/20/2022] Open
Abstract
Hypertrophic cardiomyopathy associated with damaging variants in the ALPK3 gene is a fairly recent discovery, and only a small number of patients have been described thus far. Here we present two additional patients with hypertrophic cardiomyopathy caused by biallelic variants in ALPK3. Genetic investigation was performed using a targeted gene panel consisting of known cardiomyopathy-associated genes and whole exome sequencing. The patients showed a large difference in the age of onset, and both presented with extracardiac features that are often seen in ALPK3 patients. The patient with the later onset showed milder extracardiac symptoms, such as decreased muscle tone and distal muscular dystrophy, but had fast progression of cardiac complications leading to the need of heart transplantation. This study further elucidates the variability of both symptoms and age of onset among these patients.
Collapse
Affiliation(s)
- John Jorholt
- Department of Women’s and Children’s Health and Center for Molecular Medicine, Karolinska Institute, 17177 Stockholm, Sweden; (T.S.); (A.K.)
- Correspondence:
| | - Yulia Formicheva
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia (T.V.); (A.M.); (E.D.); (P.F.); (A.Z.); (A.R.); (E.V.)
| | - Tatyana Vershinina
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia (T.V.); (A.M.); (E.D.); (P.F.); (A.Z.); (A.R.); (E.V.)
| | - Artem Kiselev
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia (T.V.); (A.M.); (E.D.); (P.F.); (A.Z.); (A.R.); (E.V.)
| | - Alexey Muravyev
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia (T.V.); (A.M.); (E.D.); (P.F.); (A.Z.); (A.R.); (E.V.)
| | - Elena Demchenko
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia (T.V.); (A.M.); (E.D.); (P.F.); (A.Z.); (A.R.); (E.V.)
| | - Petr Fedotov
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia (T.V.); (A.M.); (E.D.); (P.F.); (A.Z.); (A.R.); (E.V.)
| | - Anna Zlotina
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia (T.V.); (A.M.); (E.D.); (P.F.); (A.Z.); (A.R.); (E.V.)
| | - Anton Rygkov
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia (T.V.); (A.M.); (E.D.); (P.F.); (A.Z.); (A.R.); (E.V.)
| | - Elena Vasichkina
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia (T.V.); (A.M.); (E.D.); (P.F.); (A.Z.); (A.R.); (E.V.)
| | - Thomas Sejersen
- Department of Women’s and Children’s Health and Center for Molecular Medicine, Karolinska Institute, 17177 Stockholm, Sweden; (T.S.); (A.K.)
| | - Anna Kostareva
- Department of Women’s and Children’s Health and Center for Molecular Medicine, Karolinska Institute, 17177 Stockholm, Sweden; (T.S.); (A.K.)
- Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia (T.V.); (A.M.); (E.D.); (P.F.); (A.Z.); (A.R.); (E.V.)
| |
Collapse
|
34
|
Ravenscroft G, Clayton JS, Faiz F, Sivadorai P, Milnes D, Cincotta R, Moon P, Kamien B, Edwards M, Delatycki M, Lamont PJ, Chan SH, Colley A, Ma A, Collins F, Hennington L, Zhao T, McGillivray G, Ghedia S, Chao K, O'Donnell-Luria A, Laing NG, Davis MR. Neurogenetic fetal akinesia and arthrogryposis: genetics, expanding genotype-phenotypes and functional genomics. J Med Genet 2020; 58:609-618. [PMID: 33060286 DOI: 10.1136/jmedgenet-2020-106901] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 05/16/2020] [Accepted: 07/05/2020] [Indexed: 12/19/2022]
Abstract
BACKGROUND Fetal akinesia and arthrogryposis are clinically and genetically heterogeneous and have traditionally been refractive to genetic diagnosis. The widespread availability of affordable genome-wide sequencing has facilitated accurate genetic diagnosis and gene discovery in these conditions. METHODS We performed next generation sequencing (NGS) in 190 probands with a diagnosis of arthrogryposis multiplex congenita, distal arthrogryposis, fetal akinesia deformation sequence or multiple pterygium syndrome. This sequencing was a combination of bespoke neurogenetic disease gene panels and whole exome sequencing. Only class 4 and 5 variants were reported, except for two cases where the identified variants of unknown significance (VUS) are most likely to be causative for the observed phenotype. Co-segregation studies and confirmation of variants identified by NGS were performed where possible. Functional genomics was performed as required. RESULTS Of the 190 probands, 81 received an accurate genetic diagnosis. All except two of these cases harboured class 4 and/or 5 variants based on the American College of Medical Genetics and Genomics guidelines. We identified phenotypic expansions associated with CACNA1S, CHRNB1, GMPPB and STAC3. We describe a total of 50 novel variants, including a novel missense variant in the recently identified gene for arthrogryposis with brain malformations-SMPD4. CONCLUSIONS Comprehensive gene panels give a diagnosis for a substantial proportion (42%) of fetal akinesia and arthrogryposis cases, even in an unselected cohort. Recently identified genes account for a relatively large proportion, 32%, of the diagnoses. Diagnostic-research collaboration was critical to the diagnosis and variant interpretation in many cases, facilitated genotype-phenotype expansions and reclassified VUS through functional genomics.
Collapse
Affiliation(s)
- Gina Ravenscroft
- Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia .,Faculty of Health and Medical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Joshua S Clayton
- Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia.,Faculty of Health and Medical Sciences, University of Western Australia, Nedlands, Western Australia, Australia
| | - Fathimath Faiz
- PathWest Diagnostic Genomics, Nedlands, Western Australia, Australia
| | - Padma Sivadorai
- PathWest Diagnostic Genomics, Nedlands, Western Australia, Australia
| | - Di Milnes
- Genetic Health Queensland, Royal Brisbane and Women's Hospital, Herston, Queensland, Australia
| | - Rob Cincotta
- Maternal and Fetal Medicine, Mater Mothers' Hospital, Brisbane, Queensland, Australia
| | - Phillip Moon
- Department of Obstetrics, Redland Hospital, Cleveland, Queensland, Australia
| | - Ben Kamien
- Genetic Services WA, Women and Newborn Heath Service, Subiaco, Western Australia, Australia.,Hunter Genetics, Hunter New England Health, New Lambton, New South Wales, Australia
| | - Matthew Edwards
- Hunter Genetics, Hunter New England Health, New Lambton, New South Wales, Australia
| | - Martin Delatycki
- Victorian Clinical Genetics Service, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Phillipa J Lamont
- Neurology, Royal Perth Hospital, Perth, Western Australia, Australia
| | - Sophelia Hs Chan
- Paediatric Neurology Division, Department of Paediatrics and Adolescent Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong, Hong Kong
| | - Alison Colley
- Clinical Genetics Services SWSLHD, Liverpool Hospital, Liverpool, New South Wales, Australia
| | - Alan Ma
- Department of Clinical Genetics, Children's Hospital Westmead, Sydney, New South Wales, Australia
| | - Felicity Collins
- Clinical Genetics Department, Western Sydney Genetics Program, Children's Hospitalat Westmead, Westmead, New South Wales, Australia
| | - Lucinda Hennington
- Mercy Health, Mercy Hospital for Women, Heidelberg, Victoria, Australia.,Austin Health, Melbourne, Victoria, Australia.,Alfred Health, Melbourne, Victoria, Australia
| | - Teresa Zhao
- Victorian Clinical Genetics Service, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - George McGillivray
- Victorian Clinical Genetics Service, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Sondhya Ghedia
- Department of Clinical Genetics, Royal North Shore Hospital, Sydney, New South Wales, Australia
| | - Katherine Chao
- Center for Mendelian Genomics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Anne O'Donnell-Luria
- Center for Mendelian Genomics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts, USA
| | - Nigel G Laing
- Harry Perkins Institute of Medical Research, Perth, Western Australia, Australia.,Faculty of Health and Medical Sciences, University of Western Australia, Nedlands, Western Australia, Australia.,PathWest Diagnostic Genomics, Nedlands, Western Australia, Australia
| | - Mark R Davis
- PathWest Diagnostic Genomics, Nedlands, Western Australia, Australia
| |
Collapse
|
35
|
Vershinina T, Fomicheva Y, Muravyev A, Jorholt J, Kozyreva A, Kiselev A, Gordeev M, Vasichkina E, Segrushichev A, Pervunina T, Sjoberg G, Skyttner-Rahmani S, Sejersen T, Kostareva A. Genetic Spectrum of Left Ventricular Non-Compaction in Paediatric Patients. Cardiology 2020; 145:746-756. [PMID: 33049752 DOI: 10.1159/000510439] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 07/11/2020] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Left ventricular non-compaction (LVNC) represents a genetically heterogeneous cardiomyopathy which occurs in both children and adults. Its genetic spectrum overlaps with other types of cardiomyopathy. However, LVNC phenotypes in different age groups can have distinct genetic aetiologies. The aim of the study was to decipher the genetic spectrum of LVNC presented in childhood. Patient Group and Methods: Twenty patients under the age of 18 years diagnosed with LVNC were enrolled in the study. Target sequencing and whole-exome sequencing were performed using a panel of 108 cardiomyopathy-associated genes. Pathogenic, likely pathogenic, and variants of unknown significance found in genes highly expressed in cardiomyocytes were considered as variants of interest for further analysis. RESULTS The median age at presentation was 8.0 (0.1-17) years, with 6 patients presenting before 1 year of age. Twelve (60%) patients demonstrated reduced ejection fraction. Right ventricular (RV) dilation was registered in 6 (30%), often in combination with reduced RV contractility (25%). Almost half (45%) of the patients demonstrated biventricular involvement already at disease presentation. For pathogenic and likely pathogenic variants, the positive genotyping rate was 45%, and these variants were found mainly in non-contractile structural sarcomeric genes (ACTN2, MYPN, and TTN) or in metabolic and signal transduction genes (BRAF and TAZ). Likely pathogenic TAZ variants were detected in all 5 patients suspected of having Barth syndrome. No pathogenic or likely pathogenic variants were found in genes encoding for sarcomeric contractile proteins, but variants of unknown significance were detected in 3 out of 20 patients (MYH6, MYH7, and MYLK2). In 4 patients, variants of unknown significance in ion-channel genes were detected. CONCLUSION We detected a low burden of contractile sarcomeric variants in LVNC patients presenting below the age of 18 years, with the major number of variants residing in non-contractile structural sarcomeric genes. The identification of the variants in ion-channel and related genes not previously associated with LVNC in paediatric patients requires further examination of their functional role.
Collapse
Affiliation(s)
- Tatiana Vershinina
- Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Yulia Fomicheva
- Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Alexey Muravyev
- Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - John Jorholt
- Department of Women's and Children's Health and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Alexandra Kozyreva
- Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Artem Kiselev
- Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Michail Gordeev
- Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Elena Vasichkina
- Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | | | - Tatyana Pervunina
- Almazov National Medical Research Centre, Saint Petersburg, Russian Federation
| | - Gunnar Sjoberg
- Department of Women's and Children's Health and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Sanaz Skyttner-Rahmani
- Department of Women's and Children's Health and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Thomas Sejersen
- Department of Women's and Children's Health and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anna Kostareva
- Almazov National Medical Research Centre, Saint Petersburg, Russian Federation, .,Department of Women's and Children's Health and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden,
| |
Collapse
|
36
|
Luo YB, Peng Y, Lu Y, Li Q, Duan H, Bi F, Yang H. Expanding the Clinico-Genetic Spectrum of Myofibrillar Myopathy: Experience From a Chinese Neuromuscular Center. Front Neurol 2020; 11:1014. [PMID: 33041974 PMCID: PMC7522348 DOI: 10.3389/fneur.2020.01014] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 07/31/2020] [Indexed: 12/14/2022] Open
Abstract
Background: Myofibrillar myopathy is a group of hereditary neuromuscular disorders characterized by dissolution of myofibrils and abnormal intracellular accumulation of Z disc-related proteins. We aimed to characterize the clinical, physiological, pathohistological, and genetic features of Chinese myofibrillar myopathy patients from a single neuromuscular center. Methods: A total of 18 patients were enrolled. Demographic and clinical data were collected. Laboratory investigations, electromyography, and cardiac evaluation was performed. Routine and immunohistochemistry stainings against desmin, αB-crystallin, and BAG3 of muscle specimen were carried out. Finally, next-generation sequencing panel array for genes associated with hereditary neuromuscular disorders were performed. Results: Twelve pathogenic variants in DES, BAG3, FLNC, FHL1, and TTN were identified, of which seven were novel mutations. The novel DES c.1256C>T substitution is a high frequency mutation. The combined recessively/dominantly transmitted c.19993G>T and c.107545delG mutations in TTN gene cause a limb girdle muscular dystrophy phenotype with the classical myofibrillar myopathy histological changes. Conclusions: We report for the first time that hereditary myopathy with early respiratory failure patient can have peripheral nerve and severe spine involvement. The mutation in Ig-like domain 16 of FLNC is associated with the limb girdle type of filaminopathy, and the mutation in Ig-like domain 18 with distal myopathy type. These findings expand the phenotypic and genotypic correlation spectrum of myofibrillar myopathy.
Collapse
Affiliation(s)
- Yue-Bei Luo
- Department of Neurology, Xiangya Hospital, Central South Hospital, Changsha, China
| | - Yuyao Peng
- Department of Neurology, Xiangya Hospital, Central South Hospital, Changsha, China
| | - Yuling Lu
- Department of Neurology, the First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Qiuxiang Li
- Department of Neurology, Xiangya Hospital, Central South Hospital, Changsha, China
| | - Huiqian Duan
- Department of Neurology, Xiangya Hospital, Central South Hospital, Changsha, China
| | - Fangfang Bi
- Department of Neurology, Xiangya Hospital, Central South Hospital, Changsha, China
| | - Huan Yang
- Department of Neurology, Xiangya Hospital, Central South Hospital, Changsha, China
| |
Collapse
|
37
|
Pecorari I, Mestroni L, Sbaizero O. Current Understanding of the Role of Cytoskeletal Cross-Linkers in the Onset and Development of Cardiomyopathies. Int J Mol Sci 2020; 21:E5865. [PMID: 32824180 PMCID: PMC7461563 DOI: 10.3390/ijms21165865] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 07/28/2020] [Accepted: 08/10/2020] [Indexed: 01/08/2023] Open
Abstract
Cardiomyopathies affect individuals worldwide, without regard to age, sex and ethnicity and are associated with significant morbidity and mortality. Inherited cardiomyopathies account for a relevant part of these conditions. Although progresses have been made over the years, early diagnosis and curative therapies are still challenging. Understanding the events occurring in normal and diseased cardiac cells is crucial, as they are important determinants of overall heart function. Besides chemical and molecular events, there are also structural and mechanical phenomena that require to be investigated. Cell structure and mechanics largely depend from the cytoskeleton, which is composed by filamentous proteins that can be cross-linked via accessory proteins. Alpha-actinin 2 (ACTN2), filamin C (FLNC) and dystrophin are three major actin cross-linkers that extensively contribute to the regulation of cell structure and mechanics. Hereby, we review the current understanding of the roles played by ACTN2, FLNC and dystrophin in the onset and progress of inherited cardiomyopathies. With our work, we aim to set the stage for new approaches to study the cardiomyopathies, which might reveal new therapeutic targets and broaden the panel of genes to be screened.
Collapse
Affiliation(s)
- Ilaria Pecorari
- Department of Engineering and Architecture, University of Trieste, 34127 Trieste, Italy;
| | - Luisa Mestroni
- University of Colorado Cardiovascular Institute, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA;
| | - Orfeo Sbaizero
- Department of Engineering and Architecture, University of Trieste, 34127 Trieste, Italy;
| |
Collapse
|
38
|
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
|
39
|
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
|
40
|
Cimiotti D, Fujita-Becker S, Möhner D, Smolina N, Budde H, Wies A, Morgenstern L, Gudkova A, Sejersen T, Sjöberg G, Mügge A, Nowaczyk MM, Reusch P, Pfitzer G, Stehle R, Schröder RR, Mannherz HG, Kostareva A, Jaquet K. Infantile restrictive cardiomyopathy: cTnI-R170G/W impair the interplay of sarcomeric proteins and the integrity of thin filaments. PLoS One 2020; 15:e0229227. [PMID: 32182250 PMCID: PMC7077804 DOI: 10.1371/journal.pone.0229227] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2019] [Accepted: 01/31/2020] [Indexed: 12/11/2022] Open
Abstract
TNNI3 encoding cTnI, the inhibitory subunit of the troponin complex, is the main target for mutations leading to restrictive cardiomyopathy (RCM). Here we investigate two cTnI-R170G/W amino acid replacements, identified in infantile RCM patients, which are located in the regulatory C-terminus of cTnI. The C-terminus is thought to modulate the function of the inhibitory region of cTnI. Both cTnI-R170G/W strongly enhanced the Ca2+-sensitivity of skinned fibres, as is typical for RCM-mutations. Both mutants strongly enhanced the affinity of troponin (cTn) to tropomyosin compared to wildtype cTn, whereas binding to actin was either strengthened (R170G) or weakened (R170W). Furthermore, the stability of reconstituted thin filaments was reduced as revealed by electron microscopy. Filaments containing R170G/W appeared wavy and showed breaks. Decoration of filaments with myosin subfragment S1 was normal in the presence of R170W, but was irregular with R170G. Surprisingly, both mutants did not affect the Ca2+-dependent activation of reconstituted cardiac thin filaments. In the presence of the N-terminal fragment of cardiac myosin binding protein C (cMyBPC-C0C2) cooperativity of thin filament activation was increased only when the filaments contained wildtype cTn. No effect was observed in the presence of cTn containing R170G/W. cMyBPC-C0C2 significantly reduced binding of wildtype troponin to actin/tropomyosin, but not of both mutant cTn. Moreover, we found a direct troponin/cMyBPC-C0C2 interaction using microscale thermophoresis and identified cTnI and cTnT, but not cTnC as binding partners for cMyBPC-C0C2. Only cTn containing cTnI-R170G showed a reduced affinity towards cMyBPC-C0C2. Our results suggest that the RCM cTnI variants R170G/W impair the communication between thin and thick filament proteins and destabilize thin filaments.
Collapse
Affiliation(s)
- Diana Cimiotti
- Department of Clinical Pharmacology and Molecular Cardiology, Ruhr-University of Bochum, Bochum, Germany.,Cardiology, Bergmannsheil and St. Josef Hospital, Clinics of the Ruhr-University Bochum, Bochum, Germany
| | - Setsuko Fujita-Becker
- Cryoelectron Microscopy, BioQuant, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Desirée Möhner
- Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Natalia Smolina
- Department of Molecular Biology and Genetics, Almazov Federal Medical Research Center, St. Petersburg, Russia
| | - Heidi Budde
- Department of Clinical Pharmacology and Molecular Cardiology, Ruhr-University of Bochum, Bochum, Germany.,Cardiology, Bergmannsheil and St. Josef Hospital, Clinics of the Ruhr-University Bochum, Bochum, Germany
| | - Aline Wies
- Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Lisa Morgenstern
- Department of Clinical Pharmacology and Molecular Cardiology, Ruhr-University of Bochum, Bochum, Germany.,Cardiology, Bergmannsheil and St. Josef Hospital, Clinics of the Ruhr-University Bochum, Bochum, Germany
| | - Alexandra Gudkova
- Department of Molecular Biology and Genetics, Almazov Federal Medical Research Center, St. Petersburg, Russia
| | - Thomas Sejersen
- Department of Women's and Children's Health and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Gunnar Sjöberg
- Department of Women's and Children's Health and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Andreas Mügge
- Cardiology, Bergmannsheil and St. Josef Hospital, Clinics of the Ruhr-University Bochum, Bochum, Germany
| | - Marc M Nowaczyk
- Plant Biochemistry, Faculty of Biology and Biotechnology, Ruhr-University Bochum, Bochum, Germany
| | - Peter Reusch
- Department of Clinical Pharmacology and Molecular Cardiology, Ruhr-University of Bochum, Bochum, Germany
| | | | - Robert Stehle
- Vegetative Physiology, University of Cologne, Cologne, Germany
| | - Rasmus R Schröder
- Cryoelectron Microscopy, BioQuant, Medical Faculty, University of Heidelberg, Heidelberg, Germany
| | - Hans G Mannherz
- Department of Clinical Pharmacology and Molecular Cardiology, Ruhr-University of Bochum, Bochum, Germany.,Department of Anatomy and Embryology, Medical Faculty, Ruhr-University Bochum, Bochum, Germany
| | - Anna Kostareva
- Department of Molecular Biology and Genetics, Almazov Federal Medical Research Center, St. Petersburg, Russia.,Department of Women's and Children's Health and Center for Molecular Medicine, Karolinska Institute, Stockholm, Sweden
| | - Kornelia Jaquet
- Department of Clinical Pharmacology and Molecular Cardiology, Ruhr-University of Bochum, Bochum, Germany.,Cardiology, Bergmannsheil and St. Josef Hospital, Clinics of the Ruhr-University Bochum, Bochum, Germany
| |
Collapse
|
41
|
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
|
42
|
Abstract
Arrhythmogenic cardiomyopathy is a genetic disorder characterized by the risk of life-threatening arrhythmias, myocardial dysfunction and fibrofatty replacement of myocardial tissue. Mutations in genes that encode components of desmosomes, the adhesive junctions that connect cardiomyocytes, are the predominant cause of arrhythmogenic cardiomyopathy and can be identified in about half of patients with the condition. However, the molecular mechanisms leading to myocardial destruction, remodelling and arrhythmic predisposition remain poorly understood. Through the development of animal, induced pluripotent stem cell and other models of disease, advances in our understanding of the pathogenic mechanisms of arrhythmogenic cardiomyopathy over the past decade have brought several signalling pathways into focus. These pathways include canonical and non-canonical WNT signalling, the Hippo-Yes-associated protein (YAP) pathway and transforming growth factor-β signalling. These studies have begun to identify potential therapeutic targets whose modulation has shown promise in preclinical models. In this Review, we summarize and discuss the reported molecular mechanisms underlying the pathogenesis of arrhythmogenic cardiomyopathy.
Collapse
|
43
|
Amodeo A, Filippelli S, Perri G, Iacobelli R, Adorisio R, Iodice F, Rizza A, Massicotte MP, Baldwin JT, Almond CS. First human implantation of a miniaturized axial flow ventricular assist device in a child with end-stage heart failure. J Heart Lung Transplant 2020; 39:83-87. [DOI: 10.1016/j.healun.2019.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 07/09/2019] [Accepted: 09/03/2019] [Indexed: 10/26/2022] Open
|
44
|
Brodehl A, Ebbinghaus H, Deutsch MA, Gummert J, Gärtner A, Ratnavadivel S, Milting H. Human Induced Pluripotent Stem-Cell-Derived Cardiomyocytes as Models for Genetic Cardiomyopathies. Int J Mol Sci 2019; 20:ijms20184381. [PMID: 31489928 PMCID: PMC6770343 DOI: 10.3390/ijms20184381] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 08/29/2019] [Accepted: 09/03/2019] [Indexed: 12/17/2022] Open
Abstract
In the last few decades, many pathogenic or likely pathogenic genetic mutations in over hundred different genes have been described for non-ischemic, genetic cardiomyopathies. However, the functional knowledge about most of these mutations is still limited because the generation of adequate animal models is time-consuming and challenging. Therefore, human induced pluripotent stem cells (iPSCs) carrying specific cardiomyopathy-associated mutations are a promising alternative. Since the original discovery that pluripotency can be artificially induced by the expression of different transcription factors, various patient-specific-induced pluripotent stem cell lines have been generated to model non-ischemic, genetic cardiomyopathies in vitro. In this review, we describe the genetic landscape of non-ischemic, genetic cardiomyopathies and give an overview about different human iPSC lines, which have been developed for the disease modeling of inherited cardiomyopathies. We summarize different methods and protocols for the general differentiation of human iPSCs into cardiomyocytes. In addition, we describe methods and technologies to investigate functionally human iPSC-derived cardiomyocytes. Furthermore, we summarize novel genome editing approaches for the genetic manipulation of human iPSCs. This review provides an overview about the genetic landscape of inherited cardiomyopathies with a focus on iPSC technology, which might be of interest for clinicians and basic scientists interested in genetic cardiomyopathies.
Collapse
Affiliation(s)
- Andreas Brodehl
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hans Ebbinghaus
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Marcus-André Deutsch
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Jan Gummert
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
- Department of Thoracic and Cardiovascular Surgery, Heart and Diabetes Center NRW, University Hospital Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Anna Gärtner
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Sandra Ratnavadivel
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| | - Hendrik Milting
- Erich and Hanna Klessmann Institute, Heart and Diabetes Center NRW, University Hospital of the Ruhr-University Bochum, Georgstrasse 11, D-32545 Bad Oeynhausen, Germany.
| |
Collapse
|
45
|
ANO5 mutations in the Polish limb girdle muscular dystrophy patients: Effects on the protein structure. Sci Rep 2019; 9:11533. [PMID: 31395899 PMCID: PMC6687736 DOI: 10.1038/s41598-019-47849-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Accepted: 07/25/2019] [Indexed: 01/06/2023] Open
Abstract
LGMD2L is a subtype of limb-girdle muscular dystrophy (LGMD), caused by recessive mutations in ANO5, encoding anoctamin-5 (ANO5). We present the analysis of five patients with skeletal muscle weakness for whom heterozygous mutations within ANO5 were identified by whole exome sequencing (WES). Patients varied in the age of the disease onset (from 22 to 38 years) and severity of the morphological and clinical phenotypes. Out of the nine detected mutations one was novel (missense p.Lys132Met, accompanied by p.His841Asp) and one was not yet characterized in the literature (nonsense, p.Trp401Ter, accompanied by p.Asp81Gly). The p.Asp81Gly mutation was also identified in another patient carrying a p.Arg758Cys mutation as well. Also, a c.191dupA frameshift (p.Asn64LysfsTer15), the first described and common mutation was identified. Mutations were predicted by in silico tools to have damaging effects and are likely pathogenic according to criteria of the American College of Medical Genetics and Genomics (ACMG). Indeed, molecular modeling of mutations revealed substantial changes in ANO5 conformation that could affect the protein structure and function. In addition, variants in other genes associated with muscle pathology were identified, possibly affecting the disease progress. The presented data indicate that the identified ANO5 mutations contribute to the observed muscle pathology and broaden the genetic spectrum of LGMD myopathies.
Collapse
|
46
|
Kiselev A, Vaz R, Knyazeva A, Sergushichev A, Dmitrieva R, Khudiakov A, Jorholt J, Smolina N, Sukhareva K, Fomicheva Y, Mikhaylov E, Mitrofanova L, Predeus A, Sjoberg G, Rudenko D, Sejersen T, Lindstrand A, Kostareva A. Truncating Variant in Myof Gene Is Associated With Limb-Girdle Type Muscular Dystrophy and Cardiomyopathy. Front Genet 2019; 10:608. [PMID: 31297131 PMCID: PMC6607695 DOI: 10.3389/fgene.2019.00608] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Accepted: 06/11/2019] [Indexed: 11/13/2022] Open
Abstract
Even though genetic studies of individuals with neuromuscular diseases have uncovered the molecular background of many cardiac disorders such as cardiomyopathies and inherited arrhythmic syndromes, the genetic cause of a proportion of cardiomyopathies associated with neuromuscular phenotype still remains unknown. Here, we present an individual with a combination of cardiomyopathy and limb-girdle type muscular dystrophy where whole exome sequencing identified myoferlin (MYOF)-a member of the Ferlin protein family and close homolog of DYSF-as the most likely candidate gene. The disease-causative role of the identified variant c.[2576delG; 2575G>C], p.G859QfsTer8 is supported by functional studies in vitro using the primary patient's skeletal muscle mesenchymal progenitor cells, including both RNA sequencing and morphological studies, as well as recapitulating the muscle phenotype in vivo in zebrafish. We provide the first evidence supporting a role of MYOF in human muscle disease.
Collapse
Affiliation(s)
- Artem Kiselev
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Raquel Vaz
- Department of Molecular Medicine and Surgery, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Anastasia Knyazeva
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | | | - Renata Dmitrieva
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Aleksandr Khudiakov
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - John Jorholt
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Natalia Smolina
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Ksenia Sukhareva
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Yulia Fomicheva
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Evgeny Mikhaylov
- Arrhythmia Department, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Lubov Mitrofanova
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Alexander Predeus
- Computer Technologies Laboratory, ITMO University, Saint Petersburg, Russia.,Bioinformatics Institute, Saint Petersburg, Russia
| | - Gunnar Sjoberg
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Dmitriy Rudenko
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Thomas Sejersen
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Anna Lindstrand
- Department of Molecular Medicine and Surgery, Centre for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden.,Clinical Genetics, Karolinska University Laboratory, Karolinska University Hospital, Stockholm, Sweden
| | - Anna Kostareva
- Department of Molecular Biology and Genetics, Almazov National Medical Research Centre, Saint Petersburg, Russia.,Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
47
|
Bains S, Tester DJ, Asirvatham SJ, Noseworthy PA, Ackerman MJ, Giudicessi JR. A Novel Truncating Variant in FLNC-Encoded Filamin C May Serve as a Proarrhythmic Genetic Substrate for Arrhythmogenic Bileaflet Mitral Valve Prolapse Syndrome. Mayo Clin Proc 2019; 94:906-913. [PMID: 30935706 DOI: 10.1016/j.mayocp.2018.11.028] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Revised: 10/29/2018] [Accepted: 11/13/2018] [Indexed: 11/23/2022]
Abstract
A 51-year-old man with a long-standing history of bileaflet mitral valve prolapse accompanied by mitral annular disjunction and mild mitral regurgitation presented for evaluation of increasingly frequent palpitations. Ambulatory Holter monitoring, cardiac magnetic resonance imaging, serial transthoracic echocardiography, and diagnostic electrophysiology studies were consistent with a diagnosis of arrhythmogenic bileaflet mitral valve prolapse syndrome. Because of the presence of a similar phenotype in the proband's mother, brother, and maternal aunt, research-based whole exome sequencing was pursued and a novel truncating variant (p.Trp34*-FLNC) in the cardiomyopathy-causative FLNC-encoded filamin C unearthed that cosegregated with disease. Unexpectedly, these observations provide the first evidence that a heritable proarrhythmic genetic substrate (ie, FLNC haploinsufficiency-mediated weakening of cell-cell adhesion) may underlie, at least in part, some cases of arrhythmogenic mitral valve prolapse syndrome.
Collapse
Affiliation(s)
- Sahej Bains
- Mayo Clinic Medical Scientist Training Program, Mayo Clinic, Rochester, MN
| | - David J Tester
- Division of Pediatric Cardiology, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Molecular Pharmacology and Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN
| | - Samuel J Asirvatham
- Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Peter A Noseworthy
- Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - Michael J Ackerman
- Division of Pediatric Cardiology, Department of Pediatrics, Mayo Clinic, Rochester, MN; Department of Molecular Pharmacology and Experimental Therapeutics, Windland Smith Rice Sudden Death Genomics Laboratory, Mayo Clinic, Rochester, MN; Division of Heart Rhythm Services, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN
| | - John R Giudicessi
- Clinician-Investigator Training Program, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN.
| |
Collapse
|
48
|
Ravenscroft G, Bryson-Richardson RJ, Nowak KJ, Laing NG. Recent advances in understanding congenital myopathies. F1000Res 2018; 7. [PMID: 30631434 PMCID: PMC6290972 DOI: 10.12688/f1000research.16422.1] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2018] [Indexed: 12/18/2022] Open
Abstract
By definition, congenital myopathy typically presents with skeletal muscle weakness and hypotonia at birth. Traditionally, congenital myopathy subtypes have been predominantly distinguished on the basis of the pathological hallmarks present on skeletal muscle biopsies. Many genes cause congenital myopathies when mutated, and a burst of new causative genes have been identified because of advances in gene sequencing technology. Recent discoveries include extending the disease phenotypes associated with previously identified genes and determining that genes formerly known to cause only dominant disease can also cause recessive disease. The more recently identified congenital myopathy genes account for only a small proportion of patients. Thus, the congenital myopathy genes remaining to be discovered are predicted to be extremely rare causes of disease, which greatly hampers their identification. Significant progress in the provision of molecular diagnoses brings important information and value to patients and their families, such as possible disease prognosis, better disease management, and informed reproductive choice, including carrier screening of parents. Additionally, from accurate genetic knowledge, rational treatment options can be hypothesised and subsequently evaluated
in vitro and in animal models. A wide range of potential congenital myopathy therapies have been investigated on the basis of improved understanding of disease pathomechanisms, and some therapies are in clinical trials. Although large hurdles remain, promise exists for translating treatment benefits from preclinical models to patients with congenital myopathy, including harnessing proven successes for other genetic diseases.
Collapse
Affiliation(s)
- Gianina Ravenscroft
- Centre for Medical Research, The University of Western Australia, Perth, WA, Australia.,Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia
| | | | - Kristen J Nowak
- Centre for Medical Research, The University of Western Australia, Perth, WA, Australia.,Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,School of Biological Sciences, Faculty of Health and Medical Sciences, The University of Western Australia, QEII Medical Centre, Nedlands, WA, Australia.,Office of Population Health Genomics, Western Australian Department of Health, East Perth, WA, Australia
| | - Nigel G Laing
- Centre for Medical Research, The University of Western Australia, Perth, WA, Australia.,Harry Perkins Institute of Medical Research, QEII Medical Centre, Nedlands, WA, Australia.,Department of Diagnostic Genomics, PathWest Laboratory Medicine, QEII Medical Centre, Nedlands, WA, Australia
| |
Collapse
|
49
|
Dong J, Williams N, Cerrone M, Borck C, Wang D, Zhou B, Eng LS, Subbotina E, Um SY, Lin Y, Ruiter K, Rojas L, Coetzee WA, Sampson BA, Tang Y. Molecular autopsy: using the discovery of a novel de novo pathogenic variant in the KCNH2 gene to inform healthcare of surviving family. Heliyon 2018; 4:e01015. [PMID: 30582040 PMCID: PMC6288419 DOI: 10.1016/j.heliyon.2018.e01015] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/24/2018] [Accepted: 12/04/2018] [Indexed: 01/23/2023] Open
Abstract
Background Molecular testing of the deceased (Molecular Autopsy) is an overlooked area in the United States healthcare system and is not covered by medical insurance, leading to ineffective care for surviving families of thousands of sudden unexpected natural deaths each year. We demonstrated the precision management of surviving family members through the discovery of a novel de novo pathogenic variant in a decedent. Methods Forensic investigation and molecular autopsy were performed on an 18-year-old female who died suddenly and unexpectedly. Co-segregation family study of the first-degree relatives and functional characterization of the variant were conducted. Findings We identified a novel nonsense variant, NP_000229.1:p.Gln1068Ter, in the long QT syndrome type II gene KCNH2 in the decedent. This finding correlated with her ante-mortem electrocardiograms. Patch clamp functional studies using transfected COS-7 cells show that hERG-ΔQ1068 has a mixed phenotype, with both gain- (negative voltage shift of steady-state activation curve, the positive shift of the steady-state inactivation curve, and accelerated activation) and loss-of function (reduced current density, reduced surface expression and accelerated deactivation) hallmarks. Loss of cumulative activation during rapid pacing demonstrates that the loss-of-function phenotype predominates. The wild-type channel did not rescue the hERG-ΔQ1068 defects, demonstrating haploinsufficiency of the heterozygous state. Targeted variant testing in the family showed that the variant in KCNH2 arose de novo, which eliminated the need for exhaustive genome testing and annual cardiac follow-up for the parents and four siblings. Interpretation Molecular testing enables accurate determination of natural causes of death and precision care of the surviving family members in a time and cost-saving manner. We advocate for molecular autopsy being included under the healthcare coverage in US.
Collapse
Affiliation(s)
- Jingyun Dong
- Department of Pediatrics, NYU School of Medicine, USA.,Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, USA.,Department of Physiology and Neurosciences, NYU School of Medicine, USA
| | - Nori Williams
- Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner, USA
| | - Marina Cerrone
- Cardiovascular Genetics Program, Leon H Charney Division of Cardiology, NYU School of Medicine, NY, USA
| | - Christopher Borck
- Department of Pathology, New York City Office of Chief Medical Examiner, USA
| | - Dawei Wang
- Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner, USA
| | - Bo Zhou
- Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner, USA
| | - Lucy S Eng
- Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner, USA
| | - Ekaterina Subbotina
- Department of Pediatrics, NYU School of Medicine, USA.,Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, USA.,Department of Physiology and Neurosciences, NYU School of Medicine, USA
| | - Sung Yon Um
- Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner, USA
| | - Ying Lin
- Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner, USA
| | - Kevin Ruiter
- Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner, USA
| | - Lisa Rojas
- Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner, USA
| | - William A Coetzee
- Department of Pediatrics, NYU School of Medicine, USA.,Department of Biochemistry and Molecular Pharmacology, NYU School of Medicine, USA.,Department of Physiology and Neurosciences, NYU School of Medicine, USA
| | - Barbara A Sampson
- Department of Pathology, New York City Office of Chief Medical Examiner, USA
| | - Yingying Tang
- Molecular Genetics Laboratory, New York City Office of Chief Medical Examiner, USA
| |
Collapse
|
50
|
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
|