1
|
Darras BT, Volpe JJ. Muscle Involvement and Restricted Disorders. VOLPE'S NEUROLOGY OF THE NEWBORN 2025:1074-1121.e18. [DOI: 10.1016/b978-0-443-10513-5.00037-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
2
|
van Kleef ESB, van de Camp SAJH, Groothuis JT, Erasmus CE, Gaytant MA, Vosse BAH, de Weerd W, Verschuuren-Bemelmans CC, Medici-Van den Herik EG, Wallgren-Pettersson C, Küsters B, Schouten M, van Engelen BGM, Ottenheijm CAC, Doorduin J, Voermans NC. A cross-sectional study in 18 patients with typical and mild forms of nemaline myopathy in the Netherlands. Neuromuscul Disord 2024; 43:29-38. [PMID: 39180840 DOI: 10.1016/j.nmd.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 07/15/2024] [Accepted: 08/01/2024] [Indexed: 08/27/2024]
Abstract
Nemaline myopathy (NM) is a congenital myopathy with generalised muscle weakness, most pronounced in neck flexor, bulbar and respiratory muscles. The aim of this cross-sectional study was to assess the Dutch NM patient cohort. We assessed medical history, physical examination, quality of life (QoL), fatigue severity, motor function (MFM), and respiratory muscle function. We included 18 of the 28 identified patients (13 females (11-67 years old); five males (31-74 years old)) with typical or mild NM and eight different genotypes. Nine patients (50 %) used a wheelchair, eight patients (44 %) used mechanical ventilation, and four patients (22 %) were on tube feeding. Spinal deformities were found in 14 patients (78 %). The median Medical Research Council (MRC) sum score was 38/60 [interquartile range 32-51] in typical and 48/60 [44-50] in mild NM. The experienced QoL was lower and fatigue severity was higher than reference values of the healthy population. The total MFM score was 55 % [49-94] in typical and 88 % [72-93] in mild NM. Most of the patients who performed spirometry had a restrictive lung function pattern (11/15). This identification and characterisation of the Dutch NM patient cohort is important for international collaboration and can guide the design of future clinical trials.
Collapse
Affiliation(s)
- Esmee S B van Kleef
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud university Medical Center, Geert Grooteplein Zuid 10, Nijmegen 6525 GA, the Netherlands
| | - Sanne A J H van de Camp
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud university Medical Center, Geert Grooteplein Zuid 10, Nijmegen 6525 GA, the Netherlands
| | - Jan T Groothuis
- Department of Rehabilitation, Donders Institute for Brain, Cognition and Behaviour, Radboud university Medical Center, Nijmegen, the Netherlands
| | - Corrie E Erasmus
- Department of Paediatric Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center- Amalia Children's Hospital, Nijmegen, the Netherlands
| | - Michael A Gaytant
- Center for Home Mechanical Ventilation, Department of Pulmonology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Bettine A H Vosse
- Department of Pulmonary Diseases, Maastricht University Medical Center, Maastricht, the Netherlands
| | - Willemien de Weerd
- Department of Pediatrics, Beatrix Children's Hospital, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Evita G Medici-Van den Herik
- Department of Paediatric Neurology Erasmus MC- Sophia Children's Hospital, University Medical Center Rotterdam, the Netherlands
| | - Carina Wallgren-Pettersson
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Helsinki, Finland; Department of Medical and Clinical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Benno Küsters
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Meyke Schouten
- Department of Genetics, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Baziel G M van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud university Medical Center, Geert Grooteplein Zuid 10, Nijmegen 6525 GA, the Netherlands
| | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam, the Netherlands
| | - Jonne Doorduin
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud university Medical Center, Geert Grooteplein Zuid 10, Nijmegen 6525 GA, the Netherlands
| | - Nicol C Voermans
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud university Medical Center, Geert Grooteplein Zuid 10, Nijmegen 6525 GA, the Netherlands.
| |
Collapse
|
3
|
Iwanski JB, Pappas CT, Mayfield RM, Farman GP, Ahrens-Nicklas R, Churko JM, Gregorio CC. Leiomodin 2 neonatal dilated cardiomyopathy mutation results in altered actin gene signatures and cardiomyocyte dysfunction. NPJ Regen Med 2024; 9:21. [PMID: 39285234 PMCID: PMC11405699 DOI: 10.1038/s41536-024-00366-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 08/23/2024] [Indexed: 09/19/2024] Open
Abstract
Neonatal dilated cardiomyopathy (DCM) is a poorly understood muscular disease of the heart. Several homozygous biallelic variants in LMOD2, the gene encoding the actin-binding protein Leiomodin 2, have been identified to result in severe DCM. Collectively, LMOD2-related cardiomyopathies present with cardiac dilation and decreased heart contractility, often resulting in neonatal death. Thus, it is evident that Lmod2 is essential to normal human cardiac muscle function. This study aimed to understand the underlying pathophysiology and signaling pathways related to the first reported LMOD2 variant (c.1193 G > A, p.Trp398*). Using patient-specific human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) and a mouse model harboring the homologous mutation to the patient, we discovered dysregulated actin-thin filament lengths, altered contractility and calcium handling properties, as well as alterations in the serum response factor (SRF)-dependent signaling pathway. These findings reveal that LMOD2 may be regulating SRF activity in an actin-dependent manner and provide a potential new strategy for the development of biologically active molecules to target LMOD2-related cardiomyopathies.
Collapse
Grants
- R01HL123078 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- R00 HL128906 NHLBI NIH HHS
- R01 HL164644 NHLBI NIH HHS
- R01 GM120137 NIGMS NIH HHS
- F30HL151139 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32HL007249 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- T32 HL007249 NHLBI NIH HHS
- R01 HL123078 NHLBI NIH HHS
- R01HL164644 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- F30 HL151139 NHLBI NIH HHS
- R01GM120137 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
Collapse
Affiliation(s)
- Jessika B Iwanski
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Christopher T Pappas
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Rachel M Mayfield
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Gerrie P Farman
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA
| | - Rebecca Ahrens-Nicklas
- Department of Pediatrics and Division of Human Genetics and Metabolism, The Children's Hospital of Philadelphia, Philadelphia, PA, 19104, USA
| | - Jared M Churko
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA.
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, AZ, 85724, USA.
- Department of Medicine and Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
| |
Collapse
|
4
|
Pappas CT, Mayfield RM, Dickerson AE, Mi-Mi L, Gregorio CC. Human disease-causing mutations result in loss of leiomodin 2 through nonsense-mediated mRNA decay. PLoS Genet 2024; 20:e1011279. [PMID: 38748723 PMCID: PMC11132695 DOI: 10.1371/journal.pgen.1011279] [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: 10/02/2023] [Revised: 05/28/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
The leiomodin (Lmod) family of actin-binding proteins play a critical role in muscle function, highlighted by the fact that mutations in all three family members (LMOD1-3) result in human myopathies. Mutations in the cardiac predominant isoform, LMOD2 lead to severe neonatal dilated cardiomyopathy. Most of the disease-causing mutations in the LMOD gene family are nonsense, or frameshift, mutations predicted to result in expression of truncated proteins. However, in nearly all cases of disease, little to no LMOD protein is expressed. We show here that nonsense-mediated mRNA decay, a cellular mechanism which eliminates mRNAs with premature termination codons, underlies loss of mutant protein from two independent LMOD2 disease-causing mutations. Furthermore, we generated steric-blocking oligonucleotides that obstruct deposition of the exon junction complex, preventing nonsense-mediated mRNA decay of mutant LMOD2 transcripts, thereby restoring mutant protein expression. Our investigation lays the initial groundwork for potential therapeutic intervention in LMOD-linked myopathies.
Collapse
Affiliation(s)
- Christopher T. Pappas
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States of America
| | - Rachel M. Mayfield
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States of America
| | - Ava E. Dickerson
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States of America
| | - Lei Mi-Mi
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States of America
| | - Carol C. Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, University of Arizona, Tucson, Arizona, United States of America
- Department of Medicine and Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York City, New York, United States of America
| |
Collapse
|
5
|
Marchant RG, Bryen SJ, Bahlo M, Cairns A, Chao KR, Corbett A, Davis MR, Ganesh VS, Ghaoui R, Jones KJ, Kornberg AJ, Lek M, Liang C, MacArthur DG, Oates EC, O'Donnell-Luria A, O'Grady GL, Osei-Owusu IA, Rafehi H, Reddel SW, Roxburgh RH, Ryan MM, Sandaradura SA, Scott LW, Valkanas E, Weisburd B, Young H, Evesson FJ, Waddell LB, Cooper ST. Genome and RNA sequencing boost neuromuscular diagnoses to 62% from 34% with exome sequencing alone. Ann Clin Transl Neurol 2024; 11:1250-1266. [PMID: 38544359 DOI: 10.1002/acn3.52041] [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: 02/02/2024] [Accepted: 02/24/2024] [Indexed: 05/15/2024] Open
Abstract
OBJECTIVE Most families with heritable neuromuscular disorders do not receive a molecular diagnosis. Here we evaluate diagnostic utility of exome, genome, RNA sequencing, and protein studies and provide evidence-based recommendations for their integration into practice. METHODS In total, 247 families with suspected monogenic neuromuscular disorders who remained without a genetic diagnosis after standard diagnostic investigations underwent research-led massively parallel sequencing: neuromuscular disorder gene panel, exome, genome, and/or RNA sequencing to identify causal variants. Protein and RNA studies were also deployed when required. RESULTS Integration of exome sequencing and auxiliary genome, RNA and/or protein studies identified causal or likely causal variants in 62% (152 out of 247) of families. Exome sequencing alone informed 55% (83 out of 152) of diagnoses, with remaining diagnoses (45%; 69 out of 152) requiring genome sequencing, RNA and/or protein studies to identify variants and/or support pathogenicity. Arrestingly, novel disease genes accounted for <4% (6 out of 152) of diagnoses while 36.2% of solved families (55 out of 152) harbored at least one splice-altering or structural variant in a known neuromuscular disorder gene. We posit that contemporary neuromuscular disorder gene-panel sequencing could likely provide 66% (100 out of 152) of our diagnoses today. INTERPRETATION Our results emphasize thorough clinical phenotyping to enable deep scrutiny of all rare genetic variation in phenotypically consistent genes. Post-exome auxiliary investigations extended our diagnostic yield by 81% overall (34-62%). We present a diagnostic algorithm that details deployment of genomic and auxiliary investigations to obtain these diagnoses today most effectively. We hope this provides a practical guide for clinicians as they gain greater access to clinical genome and transcriptome sequencing.
Collapse
Affiliation(s)
- Rhett G Marchant
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Samantha J Bryen
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Melanie Bahlo
- Functional Neuromics, Children's Medical Research Institute, Westmead, New South Wales, Australia
- Population Health and Immunity, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Anita Cairns
- Department of Medical Biology, The University of Melbourne, Parkville, Victoria, Australia
- Neurosciences Department, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Katherine R Chao
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Alastair Corbett
- Neurology Department, Repatriation General Hospital Concord, Concord, New South Wales, Australia
| | - Mark R Davis
- Department of Diagnostic Genomics, PathWest Laboratory Medicine, Perth, WA, Australia
| | - Vijay S Ganesh
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Neuromuscular Division, Department of Neurology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
| | - Roula Ghaoui
- Department of Neurology, Central Adelaide Local Health Network/Royal Adelaide Hospital, Adelaide, South Australia, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia
- Department of Genetics & Molecular Pathology, SA Pathology, Adelaide, South Australia, Australia
| | - Kristi J Jones
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Clinical Genetics, Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Andrew J Kornberg
- Department of Neurology, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Monkol Lek
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christina Liang
- Department of Neurology, Royal North Shore Hospital, St Leonards, New South Wales, Australia
- Neurogenetics, Northern Clinical School, Kolling Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Daniel G MacArthur
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Centre for Population Genomics, Garvan Institute of Medical Research/University of New South Wales, Sydney, New South Wales, Australia
- Centre for Population Genomics, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Emily C Oates
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Randwick, New South Wales, Australia
| | - Anne O'Donnell-Luria
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Gina L O'Grady
- Starship Children's Health, Auckland District Health Board, Auckland, New Zealand
| | - Ikeoluwa A Osei-Owusu
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Haloom Rafehi
- Functional Neuromics, Children's Medical Research Institute, Westmead, New South Wales, Australia
- Population Health and Immunity, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Stephen W Reddel
- Neurology Department, Repatriation General Hospital Concord, Concord, New South Wales, Australia
- Brain and Mind Centre, The University of Sydney, Sydney, New South Wales, Australia
| | - Richard H Roxburgh
- Department of Neurology, Auckland District Health Board, Auckland, New Zealand
- Centre of Brain Research Neurogenetics Research Clinic, University of Auckland, Auckland, New Zealand
| | - Monique M Ryan
- Department of Neurology, Royal Children's Hospital Melbourne, Parkville, Victoria, Australia
- Department of Paediatrics, University of Melbourne, Parkville, Victoria, Australia
- Neurosciences Group, Murdoch Children's Research Institute, Melbourne, Victoria, Australia
| | - Sarah A Sandaradura
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Clinical Genetics, Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Liam W Scott
- Functional Neuromics, Children's Medical Research Institute, Westmead, New South Wales, Australia
- Population Health and Immunity, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Elise Valkanas
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, Massachusetts, USA
| | - Ben Weisburd
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Helen Young
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Department of Neurology, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Paediatrics, Royal North Shore Hospital, St Leonards, New South Wales, Australia
| | - Frances J Evesson
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Leigh B Waddell
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
| | - Sandra T Cooper
- Faculty of Medicine and Health, The University of Sydney, Westmead, New South Wales, Australia
- Kids Neuroscience Centre, Kids Research, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
6
|
Karimi E, Gohlke J, van der Borgh M, Lindqvist J, Hourani Z, Kolb J, Cossette S, Lawlor MW, Ottenheijm C, Granzier H. Characterization of NEB pathogenic variants in patients reveals novel nemaline myopathy disease mechanisms and omecamtiv mecarbil force effects. Acta Neuropathol 2024; 147:72. [PMID: 38634969 PMCID: PMC11026289 DOI: 10.1007/s00401-024-02726-w] [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: 12/22/2023] [Revised: 03/19/2024] [Accepted: 03/26/2024] [Indexed: 04/19/2024]
Abstract
Nebulin, a critical protein of the skeletal muscle thin filament, plays important roles in physiological processes such as regulating thin filament length (TFL), cross-bridge cycling, and myofibril alignment. Pathogenic variants in the nebulin gene (NEB) cause NEB-based nemaline myopathy (NEM2), a genetically heterogeneous disorder characterized by hypotonia and muscle weakness, currently lacking curative therapies. In this study, we examined a cohort of ten NEM2 patients, each with unique pathogenic variants, aiming to understand their impact on mRNA, protein, and functional levels. Results show that pathogenic truncation variants affect NEB mRNA stability and lead to nonsense-mediated decay of the mutated transcript. Moreover, a high incidence of cryptic splice site activation was found in patients with pathogenic splicing variants that are expected to disrupt the actin-binding sites of nebulin. Determination of protein levels revealed patients with either relatively normal or markedly reduced nebulin. We observed a positive relation between the reduction in nebulin and a reduction in TFL, or reduction in tension (both maximal and submaximal tension). Interestingly, our study revealed a pathogenic duplication variant in nebulin that resulted in a four-copy gain in the triplicate region of NEB and a much larger nebulin protein and longer TFL. Additionally, we investigated the effect of Omecamtiv mecarbil (OM), a small-molecule activator of cardiac myosin, on force production of type 1 muscle fibers of NEM2 patients. OM treatment substantially increased submaximal tension across all NEM2 patients ranging from 87 to 318%, with the largest effects in patients with the lowest level of nebulin. In summary, this study indicates that post-transcriptional or post-translational mechanisms regulate nebulin expression. Moreover, we propose that the pathomechanism of NEM2 involves not only shortened but also elongated thin filaments, along with the disruption of actin-binding sites resulting from pathogenic splicing variants. Significantly, our findings highlight the potential of OM treatment to improve skeletal muscle function in NEM2 patients, especially those with large reductions in nebulin levels.
Collapse
Affiliation(s)
- Esmat Karimi
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Jochen Gohlke
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Mila van der Borgh
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Johan Lindqvist
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Zaynab Hourani
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Justin Kolb
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
| | - Stacy Cossette
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Michael W Lawlor
- Department of Pathology, Medical College of Wisconsin, Milwaukee, WI, USA
- Diverge Translational Science Laboratory, Milwaukee, WI, USA
| | - Coen Ottenheijm
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA
- Department of Physiology, Amsterdam UMC (Location VUMC), Amsterdam, Netherlands
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
7
|
Larrinaga TM, Farman GP, Mayfield RM, Yuen M, Ahrens-Nicklas RC, Cooper ST, Pappas CT, Gregorio CC. Lmod2 is necessary for effective skeletal muscle contraction. SCIENCE ADVANCES 2024; 10:eadk1890. [PMID: 38478604 PMCID: PMC10936868 DOI: 10.1126/sciadv.adk1890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 02/06/2024] [Indexed: 03/17/2024]
Abstract
Muscle contraction is a regulated process driven by the sliding of actin-thin filaments over myosin-thick filaments. Lmod2 is an actin filament length regulator and essential for life since human mutations and complete loss of Lmod2 in mice lead to dilated cardiomyopathy and death. To study the little-known role of Lmod2 in skeletal muscle, we created a mouse model with Lmod2 expressed exclusively in the heart but absent in skeletal muscle. Loss of Lmod2 in skeletal muscle results in decreased force production in fast- and slow-twitch muscles. Soleus muscle from rescued Lmod2 knockout mice have shorter thin filaments, increased Lmod3 levels, and present with a myosin fiber type switch from fast myosin heavy chain (MHC) IIA to the slower MHC I isoform. Since Lmod2 regulates thin-filament length in slow-twitch but not fast-twitch skeletal muscle and force deficits were observed in both muscle types, this work demonstrates that Lmod2 regulates skeletal muscle contraction, independent of its role in thin-filament length regulation.
Collapse
Affiliation(s)
- Tania M. Larrinaga
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ 85724, USA
| | - Gerrie P. Farman
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ 85724, USA
| | - Rachel M. Mayfield
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ 85724, USA
| | - Michaela Yuen
- Kids Neuroscience Centre, Kids Research, The Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- The Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead, NSW 2145, Australia
| | | | - Sandra T. Cooper
- Kids Neuroscience Centre, Kids Research, The Children’s Hospital at Westmead, Sydney, NSW 2145, Australia
- School of Medical Sciences, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia
- The Children’s Medical Research Institute, 214 Hawkesbury Road, Westmead, NSW 2145, Australia
| | - Christopher T. Pappas
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ 85724, USA
| | - Carol C. Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ 85724, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| |
Collapse
|
8
|
Haidong L, Yin L, Ping C, Xianzhao Z, Qi Q, Xiaoli M, Zheng L, Wenhao C, Yaguang Z, Qianqian Q. Clinico-pathological and gene features of 15 nemaline myopathy patients from a single Chinese neuromuscular center. Acta Neurol Belg 2024; 124:91-99. [PMID: 37525074 PMCID: PMC10874337 DOI: 10.1007/s13760-023-02333-8] [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: 04/17/2023] [Accepted: 07/12/2023] [Indexed: 08/02/2023]
Abstract
BACKGROUND Nemaline myopathy, the most common of the congenital myopathies, is caused by various genetic mutations. In this study, we attempted to investigate the clinical features, muscle pathology and genetic features of 15 patients with nemaline myopathy. RESULTS Among the 15 patients, there were 9 (60.00%) males and 6 (40.00%) females, and 9 (60.00%) of them came from three families respectively. The age of seeing a doctor ranged from 9 to 52 years old, the age of onset was from 5 to 23 years old, and the duration of disease ranged from 3 to 35 years. Ten out of the 15 patients had high arched palate and elongated face. Only one patient had mild respiratory muscle involvement and none had dysphagia. Muscle biopsies were performed in 9 out of the 15 patients. Pathologically, muscle fibers of different sizes, atrophic muscle fibers and compensatory hypertrophic fibers could be found, and occasionally degenerated and necrotic muscle fibers were observed. Different degrees of nemaline bodies aggregation could be seen in all 9 patients. The distribution of type I and type II muscle fibers were significantly abnormal in patients with nemaline myopathy caused by NEB gene, however, it was basically normal in patients with nemaline myopathy caused by TPM3 gene and ACTA1 gene. Electron microscopic analysis of 6 patients showed that nemaline bodies aggregated between myofibrils were found in 5(83.33%) cases, and most of them were located near the Z band, but no intranuclear rods were found. The gene analysis of 15 NM patients showed that three NM-related genes were harbored, including 11 (73.33%) patients with NEB, 3 (20.00%) patients with TPM3, and 1 (6.67%) patient with ACTA1, respectively. A total of 12 mutation sites were identified and included 10 (83.33%) mutations in exon and 2(16.67%) mutations in intron. CONCLUSIONS The clinical phenotype of nemaline myopathy is highly heterogeneous. Muscle pathology shows that nemaline bodies aggregation is an important feature for the diagnosis of NM. NEB is the most frequent causative gene in this cohort. The splicing mutation, c.21522 + 3A > G may be the hotspot mutation of the NEB gene in Chinese NM patients.
Collapse
Affiliation(s)
- Lv Haidong
- Department of Neurology, Jiaozuo People's Hospital of Henan Province, Henan, 454002, Henan Province, People's Republic of China
| | - Liu Yin
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei Province, People's Republic of China
| | - Chen Ping
- Department of Neurology, Jiaozuo People's Hospital of Henan Province, Henan, 454002, Henan Province, People's Republic of China
| | - Zheng Xianzhao
- Department of Neurology, Jiaozuo People's Hospital of Henan Province, Henan, 454002, Henan Province, People's Republic of China
| | - Qian Qi
- Department of Neurology, Jiaozuo People's Hospital of Henan Province, Henan, 454002, Henan Province, People's Republic of China
| | - Ma Xiaoli
- Department of Neurology, Jiaozuo People's Hospital of Henan Province, Henan, 454002, Henan Province, People's Republic of China
| | - Lv Zheng
- Department of Neurology, Jiaozuo People's Hospital of Henan Province, Henan, 454002, Henan Province, People's Republic of China
| | - Cui Wenhao
- Department of Neurology, Jiaozuo People's Hospital of Henan Province, Henan, 454002, Henan Province, People's Republic of China
| | - Zhou Yaguang
- Department of Neurology, Jiaozuo People's Hospital of Henan Province, Henan, 454002, Henan Province, People's Republic of China
| | - Qu Qianqian
- Department of Neurology, Jiaozuo People's Hospital of Henan Province, Henan, 454002, Henan Province, People's Republic of China.
| |
Collapse
|
9
|
Vasilescu C, Colpan M, Ojala TH, Manninen T, Mutka A, Ylänen K, Rahkonen O, Poutanen T, Martelius L, Kumari R, Hinterding H, Brilhante V, Ojanen S, Lappalainen P, Koskenvuo J, Carroll CJ, Fowler VM, Gregorio CC, Suomalainen A. Recessive TMOD1 mutation causes childhood cardiomyopathy. Commun Biol 2024; 7:7. [PMID: 38168645 PMCID: PMC10761686 DOI: 10.1038/s42003-023-05670-9] [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: 04/20/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024] Open
Abstract
Familial cardiomyopathy in pediatric stages is a poorly understood presentation of heart disease in children that is attributed to pathogenic mutations. Through exome sequencing, we report a homozygous variant in tropomodulin 1 (TMOD1; c.565C>T, p.R189W) in three individuals from two unrelated families with childhood-onset dilated and restrictive cardiomyopathy. To decipher the mechanism of pathogenicity of the R189W mutation in TMOD1, we utilized a wide array of methods, including protein analyses, biochemistry and cultured cardiomyocytes. Structural modeling revealed potential defects in the local folding of TMOD1R189W and its affinity for actin. Cardiomyocytes expressing GFP-TMOD1R189W demonstrated longer thin filaments than GFP-TMOD1wt-expressing cells, resulting in compromised filament length regulation. Furthermore, TMOD1R189W showed weakened activity in capping actin filament pointed ends, providing direct evidence for the variant's effect on actin filament length regulation. Our data indicate that the p.R189W variant in TMOD1 has altered biochemical properties and reveals a unique mechanism for childhood-onset cardiomyopathy.
Collapse
Affiliation(s)
- Catalina Vasilescu
- Research Programs Unit, Stem Cells and Metabolism, Biomedicum-Helsinki, University of Helsinki, 00290, Helsinki, Finland
| | - Mert Colpan
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, 85724, USA
| | - Tiina H Ojala
- Department of Pediatric Cardiology, Helsinki University Hospital and University of Helsinki, 00290, Helsinki, Finland
| | - Tuula Manninen
- Research Programs Unit, Stem Cells and Metabolism, Biomedicum-Helsinki, University of Helsinki, 00290, Helsinki, Finland
| | - Aino Mutka
- Department of Pathology, Helsinki University Hospital and University of Helsinki, 00290, Helsinki, Finland
| | - Kaisa Ylänen
- Tampere Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and University Hospital, 33521, Tampere, Finland
| | - Otto Rahkonen
- Department of Pediatric Cardiology, Helsinki University Hospital and University of Helsinki, 00290, Helsinki, Finland
| | - Tuija Poutanen
- Tampere Center for Child, Adolescent and Maternal Health Research, Faculty of Medicine and Health Technology, Tampere University and University Hospital, 33521, Tampere, Finland
| | - Laura Martelius
- Department of Pediatric Radiology, Helsinki University Hospital and University of Helsinki, 00290, Helsinki, Finland
| | - Reena Kumari
- HiLIFE Institute of Biotechnology, University of Helsinki, 00014, Helsinki, Finland
| | - Helena Hinterding
- Research Programs Unit, Stem Cells and Metabolism, Biomedicum-Helsinki, University of Helsinki, 00290, Helsinki, Finland
| | - Virginia Brilhante
- Research Programs Unit, Stem Cells and Metabolism, Biomedicum-Helsinki, University of Helsinki, 00290, Helsinki, Finland
| | - Simo Ojanen
- Research Programs Unit, Stem Cells and Metabolism, Biomedicum-Helsinki, University of Helsinki, 00290, Helsinki, Finland
| | - Pekka Lappalainen
- HiLIFE Institute of Biotechnology, University of Helsinki, 00014, Helsinki, Finland
| | | | - Christopher J Carroll
- Research Programs Unit, Stem Cells and Metabolism, Biomedicum-Helsinki, University of Helsinki, 00290, Helsinki, Finland
- Molecular and Clinical Sciences, St. George's, University of London, London, United Kingdom
| | - Velia M Fowler
- Department of Biological Sciences, University of Delaware, Newark, DE, 19711, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, 85724, USA.
- Cardiovascular Research Institute, Department of Medicine, Icahn School of Medicine, New York, NY, 10029, USA.
| | - Anu Suomalainen
- Research Programs Unit, Stem Cells and Metabolism, Biomedicum-Helsinki, University of Helsinki, 00290, Helsinki, Finland.
- HUSlab, Helsinki University Hospital, University of Helsinki, 00290, Helsinki, Finland.
| |
Collapse
|
10
|
Farkas D, Szikora S, Jijumon AS, Polgár TF, Patai R, Tóth MÁ, Bugyi B, Gajdos T, Bíró P, Novák T, Erdélyi M, Mihály J. Peripheral thickening of the sarcomeres and pointed end elongation of the thin filaments are both promoted by SALS and its formin interaction partners. PLoS Genet 2024; 20:e1011117. [PMID: 38198522 PMCID: PMC10805286 DOI: 10.1371/journal.pgen.1011117] [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: 06/24/2023] [Revised: 01/23/2024] [Accepted: 12/27/2023] [Indexed: 01/12/2024] Open
Abstract
During striated muscle development the first periodically repeated units appear in the premyofibrils, consisting of immature sarcomeres that must undergo a substantial growth both in length and width, to reach their final size. Here we report that, beyond its well established role in sarcomere elongation, the Sarcomere length short (SALS) protein is involved in Z-disc formation and peripheral growth of the sarcomeres. Our protein localization data and loss-of-function studies in the Drosophila indirect flight muscle strongly suggest that radial growth of the sarcomeres is initiated at the Z-disc. As to thin filament elongation, we used a powerful nanoscopy approach to reveal that SALS is subject to a major conformational change during sarcomere development, which might be critical to stop pointed end elongation in the adult muscles. In addition, we demonstrate that the roles of SALS in sarcomere elongation and radial growth are both dependent on formin type of actin assembly factors. Unexpectedly, when SALS is present in excess amounts, it promotes the formation of actin aggregates highly resembling the ones described in nemaline myopathy patients. Collectively, these findings helped to shed light on the complex mechanisms of SALS during the coordinated elongation and thickening of the sarcomeres, and resulted in the discovery of a potential nemaline myopathy model, suitable for the identification of genetic and small molecule inhibitors.
Collapse
Affiliation(s)
- Dávid Farkas
- Institute of Genetics, Biological Research Centre, Szeged, Hungary
| | - Szilárd Szikora
- Institute of Genetics, Biological Research Centre, Szeged, Hungary
| | - A. S. Jijumon
- Institute of Genetics, Biological Research Centre, Szeged, Hungary
| | - Tamás F. Polgár
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
- Doctoral School of Theoretical Medicine, University of Szeged, Szeged, Hungary
| | - Roland Patai
- Institute of Biophysics, Biological Research Centre, Szeged, Hungary
| | - Mónika Ágnes Tóth
- University of Pécs, Medical School, Department of Biophysics, Pécs, Hungary
| | - Beáta Bugyi
- University of Pécs, Medical School, Department of Biophysics, Pécs, Hungary
| | - Tamás Gajdos
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Péter Bíró
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Tibor Novák
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - Miklós Erdélyi
- Department of Optics and Quantum Electronics, University of Szeged, Szeged, Hungary
| | - József Mihály
- Institute of Genetics, Biological Research Centre, Szeged, Hungary
- University of Szeged, Department of Genetics, Szeged, Hungary
| |
Collapse
|
11
|
Pascoe JE, Zygmunt A, Ehsan Z, Gurbani N. Sleep in pediatric neuromuscular disorders. Semin Pediatr Neurol 2023; 48:101092. [PMID: 38065635 DOI: 10.1016/j.spen.2023.101092] [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: 08/14/2023] [Revised: 10/06/2023] [Accepted: 10/09/2023] [Indexed: 12/18/2023]
Abstract
Sleep disordered breathing (SDB) is prevalent among children with neuromuscular disorders (NMD). The combination of respiratory muscle weakness, altered drive, and chest wall distortion due to scoliosis make sleep a stressful state in this population. Symptomatology can range from absent to snoring, nocturnal awakenings, morning headaches, and excessive daytime sleepiness. Sequelae of untreated SDB includes cardiovascular effects, metabolic derangements, and neurocognitive concerns which can be compounded by those innate to the NMD. The clinician should have a low threshold for obtaining polysomnography and recognize the nuances of individual disorders due to disproportionately impacted muscle groups such as hypoventilation in ambulating patients from diaphragm weakness. Non-invasive or invasive ventilation are the mainstay of treatment. In this review we explore the diagnosis and treatment of SDB in children with various NMD.
Collapse
Affiliation(s)
- John E Pascoe
- Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.
| | - Alexander Zygmunt
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States; Division of Neurology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States
| | - Zarmina Ehsan
- Division of Pulmonary and Sleep Medicine, Children's Mercy-Kansas City, Kansas City, MO, United States; Department of Pediatrics, University of Missouri-Kansas City School of Medicine, Kansas City, MO, United States
| | - Neepa Gurbani
- Division of Pulmonary and Sleep Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States; Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| |
Collapse
|
12
|
Schultz LE, Colpan M, Smith GE, Mayfield RM, Larrinaga TM, Kostyukova AS, Gregorio CC. A nemaline myopathy-linked mutation inhibits the actin-regulatory functions of tropomodulin and leiomodin. Proc Natl Acad Sci U S A 2023; 120:e2315820120. [PMID: 37956287 PMCID: PMC10665800 DOI: 10.1073/pnas.2315820120] [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: 09/19/2023] [Accepted: 10/06/2023] [Indexed: 11/15/2023] Open
Abstract
Actin is a highly expressed protein in eukaryotic cells and is essential for numerous cellular processes. In particular, efficient striated muscle contraction is dependent upon the precise regulation of actin-based thin filament structure and function. Alterations in the lengths of actin-thin filaments can lead to the development of myopathies. Leiomodins and tropomodulins are members of an actin-binding protein family that fine-tune thin filament lengths, and their dysfunction is implicated in muscle diseases. An Lmod3 mutation [G326R] was previously identified in patients with nemaline myopathy (NM), a severe skeletal muscle disorder; this residue is conserved among Lmod and Tmod isoforms and resides within their homologous leucine-rich repeat (LRR) domain. We mutated this glycine to arginine in Lmod and Tmod to determine the physiological function of this residue and domain. This G-to-R substitution disrupts Lmod and Tmod's LRR domain structure, altering their binding interface with actin and destroying their abilities to regulate thin filament lengths. Additionally, this mutation renders Lmod3 nonfunctional in vivo. We found that one single amino acid is essential for folding of Lmod and Tmod LRR domains, and thus is essential for the opposing actin-regulatory functions of Lmod (filament elongation) and Tmod (filament shortening), revealing a mechanism underlying the development of NM.
Collapse
Affiliation(s)
- Lauren E. Schultz
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ85724
| | - Mert Colpan
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ85724
| | - Garry E. Smith
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA99164
| | - Rachel M. Mayfield
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ85724
| | - Tania M. Larrinaga
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ85724
| | - Alla S. Kostyukova
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA99164
| | - Carol C. Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ85724
- Department of Medicine, Cardiovascular Research Institute, Icahn School of Medicine, New York, NY10029
| |
Collapse
|
13
|
Strauss KA, Carson VJ, Bolettieri E, Everett M, Bollinger A, Bowser LE, Beiler K, Young M, Edvardson S, Fraenkel N, D'Amico A, Bertini E, Lingappa L, Chowdhury D, Lowes LP, Iammarino M, Alfano LN, Brigatti KW. WiTNNess: An international natural history study of infantile-onset TNNT1 myopathy. Ann Clin Transl Neurol 2023; 10:1972-1984. [PMID: 37632133 PMCID: PMC10647004 DOI: 10.1002/acn3.51884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 07/10/2023] [Accepted: 08/02/2023] [Indexed: 08/27/2023] Open
Abstract
OBJECTIVE We created WiTNNess as a hybrid prospective/cross-sectional observational study to simulate a clinical trial for infantile-onset TNNT1 myopathy. Our aims were to identify populations for future trial enrollment, rehearse outcome assessments, specify endpoints, and refine trial logistics. METHODS Eligible participants had biallelic pathogenic variants of TNNT1 and infantile-onset proximal weakness without confounding conditions. The primary endpoint was ventilator-free survival. "Thriving" was a secondary endpoint defined as the ability to swallow and grow normally without non-oral feeding support. Endpoints of gross motor function included independent sitting and standing as defined by the Word Health Organization, a novel TNNT1 abbreviated motor score, and video mapping of limb movement. We recorded adverse events, concomitant medications, and indices of organ function to serve as comparators of safety in future trials. RESULTS Sixteen children were enrolled in the aggregate cohort (6 prospective, 10 cross-sectional; median census age 2.3 years, range 0.5-13.8). Median ventilator-free survival was 20.2 months and probability of death or permanent mechanical ventilation was 100% by age 60 months. All six children (100%) in the prospective arm failed to thrive by age 12 months. Only 2 of 16 (13%) children in the aggregate cohort sat independently and none stood alone. Novel exploratory motor assessments also proved informative. Laboratory and imaging data suggest that primary manifestations of TNNT1 deficiency are restricted to skeletal muscle. INTERPRETATION WiTNNess allowed us to streamline and economize the collection of historical control data without compromising scientific rigor, and thereby establish a sound operational framework for future clinical trials.
Collapse
Affiliation(s)
- Kevin A. Strauss
- Clinic for Special ChildrenStrasburgPennsylvaniaUSA
- Department of PediatricsPenn Medicine‐Lancaster General HospitalPennsylvaniaLancasterUSA
- Department of PediatricsUMass Chan Medical SchoolWorcesterMassachusettsUSA
- Department of Molecular, Cell & Cancer BiologyUMass Chan Medical SchoolWorcesterMassachusettsUSA
| | - Vincent J. Carson
- Clinic for Special ChildrenStrasburgPennsylvaniaUSA
- Department of PediatricsPenn Medicine‐Lancaster General HospitalPennsylvaniaLancasterUSA
| | | | | | | | | | | | - Millie Young
- Clinic for Special ChildrenStrasburgPennsylvaniaUSA
| | - Simon Edvardson
- ALYN Hospital Pediatric and Adolescent Rehabilitation CenterJerusalemIsrael
| | - Nitay Fraenkel
- ALYN Hospital Pediatric and Adolescent Rehabilitation CenterJerusalemIsrael
| | - Adele D'Amico
- Unit of Muscular and Neurodegenerative Disorders, Department of NeurosciencesIRCCS Bambino Gesù Children's HospitalRomeItaly
| | - Enrico Bertini
- Unit of Muscular and Neurodegenerative Disorders, Department of NeurosciencesIRCCS Bambino Gesù Children's HospitalRomeItaly
| | - Lokesh Lingappa
- Department of Pediatric NeurologyRainbow Children's HospitalHyderabadIndia
| | - Devyani Chowdhury
- Cardiology Care for ChildrenLancasterPennsylvaniaUSA
- Department of CardiologyNemours Children's HealthWilmingtonDelawareUSA
| | - Linda P. Lowes
- Center for Gene TherapyNationwide Children's HospitalColumbusOhioUSA
| | - Megan Iammarino
- Center for Gene TherapyNationwide Children's HospitalColumbusOhioUSA
| | - Lindsay N. Alfano
- Center for Gene TherapyNationwide Children's HospitalColumbusOhioUSA
| | | |
Collapse
|
14
|
Hamper M, Schmidt-Kastner R. Sleep Disorder Kleine-Levin Syndrome (KLS) Joins the List of Polygenic Brain Disorders Associated with Obstetric Complications. Cell Mol Neurobiol 2023; 43:3393-3403. [PMID: 37553546 DOI: 10.1007/s10571-023-01391-z] [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: 05/15/2023] [Accepted: 07/17/2023] [Indexed: 08/10/2023]
Abstract
Kleine-Levin Syndrome is a rare neurological disorder with onset typically during adolescence that is characterized by recurrent episodes of hypersomnia, behavioral changes, and cognitive abnormalities, in the absence of structural changes in neuroimaging. As for many functional brain disorders, the exact disease mechanism in Kleine-Levin Syndrome is presently unknown, preventing the development of specific treatment approaches or protective measures. Here we review the pathophysiology and genetics of this functional brain disorder and then present a specific working hypothesis. A neurodevelopmental mechanism has been suspected based on associations with obstetric complications. Recent studies have focused on genetic factors whereby the first genome-wide association study (GWAS) in Kleine-Levin Syndrome has defined a linkage at the TRANK1 locus. A Gene x Environment interaction model involving obstetric complications was proposed based on concepts developed for other functional brain disorders. To stimulate future research, we here performed annotations of the genes under consideration for Kleine-Levin Syndrome in relation to factors expected to be associated with obstetric complications. Annotations used data-mining of gene/protein lists related to for hypoxia, ischemia, and vascular factors and targeted literature searches. Tentative links for TRANK1, four additional genes in the TRANK1 locus, and LMOD3-LMO2 are described. Protein interaction data for TRANK1 indicate links to CBX2, CBX4, and KDM3A, that in turn can be tied to hypoxia. Taken together, the neurological sleep disorder, Kleine-Levin Syndrome, shows genetic and mechanistic overlap with well analyzed brain disorders such as schizophrenia, autism spectrum disorder and ADHD in which polygenic predisposition interacts with external events during brain development, including obstetric complications.
Collapse
Affiliation(s)
- Michael Hamper
- Florida Atlantic University (FAU), CE Schmidt College of Medicine, Boca Raton, FL, USA
| | - Rainald Schmidt-Kastner
- Florida Atlantic University (FAU), CE Schmidt College of Medicine, Boca Raton, FL, USA.
- Dept. Clinical Neurosciences, CE Schmidt College of Medicine, Florida Atlantic University (FAU), 777 Glades Road, Boca Raton, FL, 33431, USA.
| |
Collapse
|
15
|
Dehghanian Reyhan V, Ghafouri F, Sadeghi M, Miraei-Ashtiani SR, Kastelic JP, Barkema HW, Shirali M. Integrated Comparative Transcriptome and circRNA-lncRNA-miRNA-mRNA ceRNA Regulatory Network Analyses Identify Molecular Mechanisms Associated with Intramuscular Fat Content in Beef Cattle. Animals (Basel) 2023; 13:2598. [PMID: 37627391 PMCID: PMC10451991 DOI: 10.3390/ani13162598] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 08/05/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
Intramuscular fat content (IMF), one of the most important carcass traits in beef cattle, is controlled by complex regulatory factors. At present, molecular mechanisms involved in regulating IMF and fat metabolism in beef cattle are not well understood. Our objective was to integrate comparative transcriptomic and competing endogenous RNA (ceRNA) network analyses to identify candidate messenger RNAs (mRNAs) and regulatory RNAs involved in molecular regulation of longissimus dorsi muscle (LDM) tissue for IMF and fat metabolism of 5 beef cattle breeds (Angus, Chinese Simmental, Luxi, Nanyang, and Shandong Black). In total, 34 circRNAs, 57 lncRNAs, 15 miRNAs, and 374 mRNAs were identified by integrating gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses. Furthermore, 7 key subnets with 16 circRNAs, 43 lncRNAs, 7 miRNAs, and 237 mRNAs were detected through clustering analyses, whereas GO enrichment analysis of identified RNAs revealed 48, 13, and 28 significantly enriched GO terms related to IMF in biological process, molecular function, and cellular component categories, respectively. The main metabolic-signaling pathways associated with IMF and fat metabolism that were enriched included metabolic, calcium, cGMP-PKG, thyroid hormone, and oxytocin signaling pathways. Moreover, MCU, CYB5R1, and BAG3 genes were common among the 10 comparative groups defined as important candidate marker genes for fat metabolism in beef cattle. Contributions of transcriptome profiles from various beef breeds and a competing endogenous RNA (ceRNA) regulatory network underlying phenotypic differences in IMF provided novel insights into molecular mechanisms associated with meat quality.
Collapse
Affiliation(s)
- Vahid Dehghanian Reyhan
- Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj 77871-31587, Iran; (V.D.R.); (F.G.); (S.R.M.-A.)
| | - Farzad Ghafouri
- Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj 77871-31587, Iran; (V.D.R.); (F.G.); (S.R.M.-A.)
| | - Mostafa Sadeghi
- Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj 77871-31587, Iran; (V.D.R.); (F.G.); (S.R.M.-A.)
| | - Seyed Reza Miraei-Ashtiani
- Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj 77871-31587, Iran; (V.D.R.); (F.G.); (S.R.M.-A.)
| | - John P. Kastelic
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (J.P.K.); (H.W.B.)
| | - Herman W. Barkema
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; (J.P.K.); (H.W.B.)
| | - Masoud Shirali
- Agri-Food and Biosciences Institute, Hillsborough BT26 6DR, UK
- School of Biological Sciences, Queen’s University Belfast, Belfast BT9 5AJ, UK
| |
Collapse
|
16
|
Casey JG, Kim ES, Joseph R, Li F, Granzier H, Gupta VA. NRAP reduction rescues sarcomere defects in nebulin-related nemaline myopathy. Hum Mol Genet 2023; 32:1711-1721. [PMID: 36661122 PMCID: PMC10162428 DOI: 10.1093/hmg/ddad011] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/18/2022] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Nemaline myopathy (NM) is a rare neuromuscular disorder associated with congenital or childhood-onset of skeletal muscle weakness and hypotonia, which results in limited motor function. NM is a genetic disorder and mutations in 12 genes are known to contribute to autosomal dominant or recessive forms of the disease. Recessive mutations in nebulin (NEB) are the most common cause of NM affecting about 50% of patients. Because of the large size of the NEB gene and lack of mutational hot spots, developing therapies that can benefit a wide group of patients is challenging. Although there are several promising therapies under investigation, there is no cure for NM. Therefore, targeting disease modifiers that can stabilize or improve skeletal muscle function may represent alternative therapeutic strategies. Our studies have identified Nrap upregulation in nebulin deficiency that contributes to structural and functional deficits in NM. We show that genetic ablation of nrap in nebulin deficiency restored sarcomeric disorganization, reduced protein aggregates and improved skeletal muscle function in zebrafish. Our findings suggest that Nrap is a disease modifier that affects skeletal muscle structure and function in NM; thus, therapeutic targeting of Nrap in nebulin-related NM and related diseases may be beneficial for patients.
Collapse
Affiliation(s)
- Jennifer G Casey
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Euri S Kim
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Remi Joseph
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Frank Li
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Henk Granzier
- Department of Cellular and Molecular Medicine, University of Arizona, Tucson, AZ 85724, USA
| | - Vandana A Gupta
- Division of Genetics, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
17
|
Zapater I Morales C, Carman PJ, Soffar DB, Windner SE, Dominguez R, Baylies MK. Drosophila Tropomodulin is required for multiple actin-dependent processes within developing myofibers. Development 2023; 150:dev201194. [PMID: 36806912 PMCID: PMC10112908 DOI: 10.1242/dev.201194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Accepted: 02/09/2023] [Indexed: 02/22/2023]
Abstract
Proper muscle contraction requires the assembly and maintenance of sarcomeres and myofibrils. Although the protein components of myofibrils are generally known, less is known about the mechanisms by which they individually function and together synergize for myofibril assembly and maintenance. For example, it is unclear how the disruption of actin filament (F-actin) regulatory proteins leads to the muscle weakness observed in myopathies. Here, we show that knockdown of Drosophila Tropomodulin (Tmod), results in several myopathy-related phenotypes, including reduction of muscle cell (myofiber) size, increased sarcomere length, disorganization and misorientation of myofibrils, ectopic F-actin accumulation, loss of tension-mediating proteins at the myotendinous junction, and misshaped and internalized nuclei. Our findings support and extend the tension-driven self-organizing myofibrillogenesis model. We show that, like its mammalian counterpart, Drosophila Tmod caps F-actin pointed-ends, and we propose that this activity is crucial for cellular processes in different locations within the myofiber that directly and indirectly contribute to the maintenance of muscle function. Our findings provide significant insights to the role of Tmod in muscle development, maintenance and disease.
Collapse
Affiliation(s)
- Carolina Zapater I Morales
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Peter J Carman
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David B Soffar
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Stefanie E Windner
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| | - Roberto Dominguez
- Biochemistry and Molecular Biophysics Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mary K Baylies
- Biochemistry, Cell & Developmental Biology, and Molecular Biology (BCMB) program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
- Developmental Biology Program, Sloan Kettering Institute, Memorial Sloan Kettering, Cancer Center, New York, NY 10065, USA
| |
Collapse
|
18
|
Segarra-Casas A, Collet R, Gonzalez-Quereda L, Vesperinas A, Caballero-Ávila M, Carbayo A, Díaz-Manera J, Rodriguez MJ, Gallardo E, Gallano P, Olivé M. A new homozygous missense variant in LMOD3 gene causing mild nemaline myopathy with prominent facial weakness. Neuromuscul Disord 2023; 33:319-323. [PMID: 36893608 DOI: 10.1016/j.nmd.2023.02.006] [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: 11/23/2022] [Revised: 01/24/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023]
Abstract
Nemaline myopathy (NEM) type 10, caused by biallelic mutations in LMOD3, is a severe congenital myopathy clinically characterized by generalized hypotonia and muscle weakness, respiratory insufficiency, joint contractures, and bulbar weakness. Here, we describe a family with two adult patients presenting mild nemaline myopathy due to a novel homozygous missense variant in LMOD3. Both patients presented mild delayed motor milestones, frequent falls during infancy, prominent facial weakness and mild muscle weakness in the four limbs. Muscle biopsy showed mild myopathic changes and small nemaline bodies in a few fibers. A neuromuscular gene panel revealed a homozygous missense variant in LMOD3 that co-segregated with the disease in the family (NM_198271.4: c.1030C>T; p.Arg344Trp). The patients described here provide evidence of the phenotype-genotype correlation, suggesting that non-truncating variants in LMOD3 lead to milder phenotypes of NEM type 10.
Collapse
Affiliation(s)
- Alba Segarra-Casas
- Genetics Department, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Genetics and Microbiology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain
| | - Roger Collet
- Neuromuscular Diseases Unit, Neurology Department, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Lidia Gonzalez-Quereda
- Genetics Department, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Genetics and Microbiology Department, Universitat Autònoma de Barcelona, Bellaterra, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Vesperinas
- Neuromuscular Diseases Unit, Neurology Department, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Marta Caballero-Ávila
- Neuromuscular Diseases Unit, Neurology Department, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Alvaro Carbayo
- Neuromuscular Diseases Unit, Neurology Department, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Jordi Díaz-Manera
- John Walton Muscular Dystrophy Research Center. Newcastle University Translational and Clinical Research Institute, Newcastle, UK
| | - María José Rodriguez
- Genetics Department, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Eduard Gallardo
- Neuromuscular Diseases Unit, Neurology Department, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Pia Gallano
- Genetics Department, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Montse Olivé
- Neuromuscular Diseases Unit, Neurology Department, Institut d'Investigació Biomèdica Sant Pau (IIB SANT PAU), Hospital de la Santa Creu i Sant Pau, Barcelona, Spain; Center for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
19
|
Pepke ML, Kvalnes T, Lundregan S, Boner W, Monaghan P, Saether BE, Jensen H, Ringsby TH. Genetic architecture and heritability of early-life telomere length in a wild passerine. Mol Ecol 2022; 31:6360-6381. [PMID: 34825754 DOI: 10.1111/mec.16288] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 10/01/2021] [Accepted: 11/09/2021] [Indexed: 01/31/2023]
Abstract
Early-life telomere length (TL) is associated with fitness in a range of organisms. Little is known about the genetic basis of variation in TL in wild animal populations, but to understand the evolutionary and ecological significance of TL it is important to quantify the relative importance of genetic and environmental variation in TL. In this study, we measured TL in 2746 house sparrow nestlings sampled across 20 years and used an animal model to show that there is a small heritable component of early-life TL (h2 = 0.04). Variation in TL among individuals was mainly driven by environmental (annual) variance, but also brood and parental effects. Parent-offspring regressions showed a large maternal inheritance component in TL ( h maternal 2 = 0.44), but no paternal inheritance. We did not find evidence for a negative genetic correlation underlying the observed negative phenotypic correlation between TL and structural body size. Thus, TL may evolve independently of body size and the negative phenotypic correlation is likely to be caused by nongenetic environmental effects. We further used genome-wide association analysis to identify genomic regions associated with TL variation. We identified several putative genes underlying TL variation; these have been inferred to be involved in oxidative stress, cellular growth, skeletal development, cell differentiation and tumorigenesis in other species. Together, our results show that TL has a low heritability and is a polygenic trait strongly affected by environmental conditions in a free-living bird.
Collapse
Affiliation(s)
- Michael Le Pepke
- Department of Biology, Centre for Biodiversity Dynamics (CBD), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Thomas Kvalnes
- Department of Biology, Centre for Biodiversity Dynamics (CBD), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sarah Lundregan
- Department of Biology, Centre for Biodiversity Dynamics (CBD), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Winnie Boner
- Institute of Biodiversity, Animal Health and Comparative Medicine (IBAHCM), University of Glasgow, Glasgow, UK
| | - Pat Monaghan
- Institute of Biodiversity, Animal Health and Comparative Medicine (IBAHCM), University of Glasgow, Glasgow, UK
| | - Bernt-Erik Saether
- Department of Biology, Centre for Biodiversity Dynamics (CBD), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Henrik Jensen
- Department of Biology, Centre for Biodiversity Dynamics (CBD), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Thor Harald Ringsby
- Department of Biology, Centre for Biodiversity Dynamics (CBD), Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
20
|
Nemaline Myopathy in Brazilian Patients: Molecular and Clinical Characterization. Int J Mol Sci 2022; 23:ijms231911995. [PMID: 36233295 PMCID: PMC9569467 DOI: 10.3390/ijms231911995] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 09/10/2022] [Accepted: 09/27/2022] [Indexed: 11/07/2022] Open
Abstract
Nemaline myopathy (NM), a structural congenital myopathy, presents a significant clinical and genetic heterogeneity. Here, we compiled molecular and clinical data of 30 Brazilian patients from 25 unrelated families. Next-generation sequencing was able to genetically classify all patients: sixteen families (64%) with mutation in NEB, five (20%) in ACTA1, two (8%) in KLHL40, and one in TPM2 (4%) and TPM3 (4%). In the NEB-related families, 25 different variants, 11 of them novel, were identified; splice site (10/25) and frame shift (9/25) mutations were the most common. Mutation c.24579 G>C was recurrent in three unrelated patients from the same region, suggesting a common ancestor. Clinically, the “typical” form was the more frequent and caused by mutations in the different NM genes. Phenotypic heterogeneity was observed among patients with mutations in the same gene. Respiratory involvement was very common and often out of proportion with limb weakness. Muscle MRI patterns showed variability within the forms and genes, which was related to the severity of the weakness. Considering the high frequency of NEB mutations and the complexity of this gene, NGS tools should be combined with CNV identification, especially in patients with a likely non-identified second mutation.
Collapse
|
21
|
Tolkatchev D, Gregorio CC, Kostyukova AS. The role of leiomodin in actin dynamics: a new road or a secret gate. FEBS J 2022; 289:6119-6131. [PMID: 34273242 PMCID: PMC8761783 DOI: 10.1111/febs.16128] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/10/2021] [Accepted: 07/16/2021] [Indexed: 12/29/2022]
Abstract
Leiomodin is an important emerging regulator of thin filaments. As novel molecular, cellular, animal model, and human data accumulate, the mechanisms of its action become clearer. Structural studies played a significant part in understanding the functional significance of leiomodin's interacting partners and functional domains. In this review, we present the current state of knowledge on the structural and cellular properties of leiomodin which has led to two proposed mechanisms of its function. Although it is known that leiomodin is essential for life, numerous domains within leiomodin remain unstudied and as such, we outline future directions for investigations that we predict will provide evidence that leiomodin is a multifunctional protein.
Collapse
Affiliation(s)
- Dmitri Tolkatchev
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164, USA
| | - Carol C. Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ 85724, USA
| | - Alla S. Kostyukova
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, WA 99164, USA
| |
Collapse
|
22
|
Fisher G, Mackels L, Markati T, Sarkozy A, Ochala J, Jungbluth H, Ramdas S, Servais L. Early clinical and pre-clinical therapy development in Nemaline myopathy. Expert Opin Ther Targets 2022; 26:853-867. [PMID: 36524401 DOI: 10.1080/14728222.2022.2157258] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Nemaline myopathies (NM) represent a group of clinically and genetically heterogeneous congenital muscle disorders with the common denominator of nemaline rods on muscle biopsy. NEB and ACTA1 are the most common causative genes. Currently, available treatments are supportive. AREAS COVERED We explored experimental treatments for NM, identifying at least eleven mainly pre-clinical approaches utilizing murine and/or human muscle cells. These approaches target either i) the causative gene or associated genes implicated in the same pathway; ii) pathophysiologically relevant biochemical mechanisms such as calcium/myosin regulation of muscle contraction; iii) myogenesis; iv) other therapies that improve or optimize muscle function more generally; v) and/or combinations of the above. The scope and efficiency of these attempts is diverse, ranging from gene-specific effects to those widely applicable to all NM-associated genes. EXPERT OPINION The wide range of experimental therapies currently under consideration for NM is promising. Potential translation into clinical use requires consideration of additional factors such as the potential muscle type specificity as well as the possibility of gene expression remodeling. Challenges in clinical translation include the rarity and heterogeneity of genotypes, phenotypes, and disease trajectories, as well as the lack of longitudinal natural history data and validated outcomes and biomarkers.
Collapse
Affiliation(s)
- Gemma Fisher
- MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Laurane Mackels
- MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, UK.,Neuromuscular Reference Center, University and University Hospital of Liège, Liège, Belgium
| | - Theodora Markati
- MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Anna Sarkozy
- Dubowitz Neuromuscular Centre, UCL Great Ormond Street Hospital, Institute of Child Health, London, UK
| | - Julien Ochala
- Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Heinz Jungbluth
- Department of Paediatric Neurology - Neuromuscular Service, Evelina Children's Hospital, Guy's & St Thomas' NHS Foundation Trust, London, UK.,Randall Centre for Cell and Molecular Biophysics, Muscle Signalling Section, Faculty of Life Sciences and Medicine (FoLSM), King's College London, London, UK
| | - Sithara Ramdas
- MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, UK.,Department of Paediatric Neurology, John Radcliffe Hospital, Oxford, UK
| | - Laurent Servais
- MDUK Neuromuscular Centre, Department of Paediatrics, University of Oxford, Oxford, UK.,Neuromuscular Reference Center, University and University Hospital of Liège, Liège, Belgium
| |
Collapse
|
23
|
Mizuta K, Katou Y, Nakakita B, Kishine A, Nosaka Y, Saito S, Iwatani C, Tsuchiya H, Kawamoto I, Nakaya M, Tsukiyama T, Nagano M, Kojima Y, Nakamura T, Yabuta Y, Horie A, Mandai M, Ohta H, Saitou M. Ex vivo reconstitution of fetal oocyte development in humans and cynomolgus monkeys. EMBO J 2022; 41:e110815. [PMID: 35912849 PMCID: PMC9475534 DOI: 10.15252/embj.2022110815] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 06/28/2022] [Accepted: 06/29/2022] [Indexed: 12/14/2022] Open
Abstract
In vitro oogenesis is key to elucidating the mechanism of human female germ-cell development and its anomalies. Accordingly, pluripotent stem cells have been induced into primordial germ cell-like cells and into oogonia with epigenetic reprogramming, yet further reconstitutions remain a challenge. Here, we demonstrate ex vivo reconstitution of fetal oocyte development in both humans and cynomolgus monkeys (Macaca fascicularis). With an optimized culture of fetal ovary reaggregates over three months, human and monkey oogonia enter and complete the first meiotic prophase to differentiate into diplotene oocytes that form primordial follicles, the source for oogenesis in adults. The cytological and transcriptomic progressions of fetal oocyte development in vitro closely recapitulate those in vivo. A comparison of single-cell transcriptomes among humans, monkeys, and mice unravels primate-specific and conserved programs driving fetal oocyte development, the former including a distinct transcriptomic transformation upon oogonia-to-oocyte transition and the latter including two active X chromosomes with little X-chromosome upregulation. Our study provides a critical step forward for realizing human in vitro oogenesis and uncovers salient characteristics of fetal oocyte development in primates.
Collapse
Affiliation(s)
- Ken Mizuta
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshitaka Katou
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Baku Nakakita
- Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Aoi Kishine
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiaki Nosaka
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Saki Saito
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Chizuru Iwatani
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Hideaki Tsuchiya
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Ikuo Kawamoto
- Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Masataka Nakaya
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Tomoyuki Tsukiyama
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Research Center for Animal Life Science, Shiga University of Medical Science, Otsu, Japan
| | - Masahiro Nagano
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoji Kojima
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Tomonori Nakamura
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,The Hakubi Center for Advanced Research, Kyoto University, Kyoto, Japan
| | - Yukihiro Yabuta
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Akihito Horie
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masaki Mandai
- Department of Gynecology and Obstetrics, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Ohta
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mitinori Saitou
- Institute for the Advanced Study of Human Biology (ASHBi), Kyoto University, Kyoto, Japan.,Department of Anatomy and Cell Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan.,Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
24
|
Smith GE, Tolkatchev D, Risi C, Little M, Gregorio CC, Galkin VE, Kostyukova AS. Ca 2+ attenuates nucleation activity of leiomodin. Protein Sci 2022; 31:e4358. [PMID: 35762710 PMCID: PMC9207750 DOI: 10.1002/pro.4358] [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: 03/02/2022] [Revised: 04/25/2022] [Accepted: 05/14/2022] [Indexed: 11/10/2022]
Abstract
A transient increase in Ca2+ concentration in sarcomeres is essential for their proper function. Ca2+ drives striated muscle contraction via binding to the troponin complex of the thin filament to activate its interaction with the myosin thick filament. In addition to the troponin complex, the myosin essential light chain and myosin-binding protein C were also found to be Ca2+ sensitive. However, the effects of Ca2+ on the function of the tropomodulin family proteins involved in regulating thin filament formation have not yet been studied. Leiomodin, a member of the tropomodulin family, is an actin nucleator and thin filament elongator. Using pyrene-actin polymerization assay and transmission electron microscopy, we show that the actin nucleation activity of leiomodin is attenuated by Ca2+ . Using circular dichroism and nuclear magnetic resonance spectroscopy, we demonstrate that the mostly disordered, negatively charged region of leiomodin located between its first two actin-binding sites binds Ca2+ . We propose that Ca2+ binding to leiomodin results in the attenuation of its nucleation activity. Our data provide further evidence regarding the role of Ca2+ as an ultimate regulator of the ensemble of sarcomeric proteins essential for muscle function. SUMMARY STATEMENT: Ca2+ fluctuations in striated muscle sarcomeres modulate contractile activity via binding to several distinct families of sarcomeric proteins. The effects of Ca2+ on the activity of leiomodin-an actin nucleator and thin filament length regulator-have remained unknown. In this study, we demonstrate that Ca2+ binds directly to leiomodin and attenuates its actin nucleating activity. Our data emphasizes the ultimate role of Ca2+ in the regulation of the sarcomeric protein interactions.
Collapse
Affiliation(s)
- Garry E. Smith
- Voiland School of Chemical Engineering and BioengineeringWashington State UniversityPullmanWashingtonUSA
| | - Dmitri Tolkatchev
- Voiland School of Chemical Engineering and BioengineeringWashington State UniversityPullmanWashingtonUSA
| | - Cristina Risi
- Department of Physiological SciencesEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Madison Little
- Voiland School of Chemical Engineering and BioengineeringWashington State UniversityPullmanWashingtonUSA
| | - Carol C. Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research ProgramUniversity of ArizonaTucsonArizonaUSA
| | - Vitold E. Galkin
- Department of Physiological SciencesEastern Virginia Medical SchoolNorfolkVirginiaUSA
| | - Alla S. Kostyukova
- Voiland School of Chemical Engineering and BioengineeringWashington State UniversityPullmanWashingtonUSA
| |
Collapse
|
25
|
Yin X, Pu C, Wang Z, Li K, Wang H. Clinico-pathological features and mutational spectrum of 16 nemaline myopathy patients from a Chinese neuromuscular center. Acta Neurol Belg 2022; 122:631-639. [PMID: 33742414 PMCID: PMC9170660 DOI: 10.1007/s13760-020-01542-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Accepted: 10/29/2020] [Indexed: 11/15/2022]
Abstract
Nemaline myopathy (NM) is a congenital myopathy of great heterogeneity, characterized by the presence of rods in the cytoplasm of muscle fibers. The samples of 16 nemaline myopathy patients diagnosed by characteristically pathological features went through whole exon sequencing. Clinico-pathological and genetic features of the cases were systematically analyzed. According to the classification of nemaline myopathy by ENMC, 8 cases are typical congenital subtype, 6 cases are childhood/juvenile onset subtype and 2 case are adult onset subtype. In histological findings, characteristic purple-colored rods are discovered under modified gömöri trichrome staining (MGT). Electron microscopy revealed the presence of high electron-dense nemaline bodies around the submucosa and the nucleus nine patients (9/16 56.3%) were detected pathogenic causative mutations, among whom mutations in the NEB gene were the most frequent (6 patients, 66.7%). KBTBD13 gene mutation was discovered in two patients and ACTA1 gene mutation was discovered in 1 patient. Nemaline myopathy is a congenital myopathy with highly clinico-pathological and genetic heterogeneity. NEB gene mutation is the most common mutation, in which splicing change c.21522 +3A > G is hotspot mutation in Chinese NM patients.
Collapse
|
26
|
Abstract
Monkol Lek, Assistant Professor at Yale University School of Medicine, and Associate Editor at Disease Models & Mechanisms, dedicates his research to finding a genetic diagnosis and improving treatments for rare disease patients. As he originally studied computer engineering at the University of New South Wales in Sydney, Australia, he now utilises computational methods to optimise large-scale genetic studies, provide globally accessible resources for genetic research communities and, importantly, resolve diagnostic odysseys for rare disease patients. Monkol completed his PhD in Prof. Kathryn North's lab at the University of Sydney, studying the genetics of muscle strength and performance, and then continued his investigation of muscle disease in Prof. Daniel MacArthur's lab at Massachusetts General Hospital and the Broad Institute. During his postdoc, he led several large-scale studies aimed at distinguishing pathogenic from benign variants, including the Exome Aggregation Consortium (ExAC) project (
Lek et al., 2016). Monkol established his own lab at Yale University School of Medicine, which continues to improve the diagnosis and treatment of rare muscle disease, and also focuses on underserved populations, whose genetic mutations are not as well characterised as those of European ancestry. In this interview, Monkol discusses how his own diagnosis with limb girdle muscular dystrophy has shaped his career and what he envisions for the future of genetic research in rare disease.
Collapse
Affiliation(s)
- Monkol Lek
- Department of Genetics, Yale University School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
27
|
van Kleef ES, van Doorn JL, Gaytant MA, de Weerd W, Vosse BA, Wallgren-Pettersson C, van Engelen BG, Ottenheijm CA, Voermans NC, Doorduin J. Respiratory muscle function in patients with nemaline myopathy. Neuromuscul Disord 2022; 32:654-663. [DOI: 10.1016/j.nmd.2022.06.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 11/24/2022]
|
28
|
Szikora S, Görög P, Mihály J. The Mechanisms of Thin Filament Assembly and Length Regulation in Muscles. Int J Mol Sci 2022; 23:5306. [PMID: 35628117 PMCID: PMC9140763 DOI: 10.3390/ijms23105306] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
The actin containing tropomyosin and troponin decorated thin filaments form one of the crucial components of the contractile apparatus in muscles. The thin filaments are organized into densely packed lattices interdigitated with myosin-based thick filaments. The crossbridge interactions between these myofilaments drive muscle contraction, and the degree of myofilament overlap is a key factor of contractile force determination. As such, the optimal length of the thin filaments is critical for efficient activity, therefore, this parameter is precisely controlled according to the workload of a given muscle. Thin filament length is thought to be regulated by two major, but only partially understood mechanisms: it is set by (i) factors that mediate the assembly of filaments from monomers and catalyze their elongation, and (ii) by factors that specify their length and uniformity. Mutations affecting these factors can alter the length of thin filaments, and in human cases, many of them are linked to debilitating diseases such as nemaline myopathy and dilated cardiomyopathy.
Collapse
Affiliation(s)
- Szilárd Szikora
- Institute of Genetics, Biological Research Centre, H-6726 Szeged, Hungary;
| | - Péter Görög
- Institute of Genetics, Biological Research Centre, H-6726 Szeged, Hungary;
- Doctoral School of Multidisciplinary Medical Science, Faculty of Medicine, University of Szeged, H-6725 Szeged, Hungary
| | - József Mihály
- Institute of Genetics, Biological Research Centre, H-6726 Szeged, Hungary;
- Department of Genetics, University of Szeged, H-6726 Szeged, Hungary
| |
Collapse
|
29
|
Yuen M, Worgan L, Iwanski J, Pappas CT, Joshi H, Churko JM, Arbuckle S, Kirk EP, Zhu Y, Roscioli T, Gregorio CC, Cooper ST. Neonatal-lethal dilated cardiomyopathy due to a homozygous LMOD2 donor splice-site variant. Eur J Hum Genet 2022; 30:450-457. [PMID: 35082396 PMCID: PMC8989920 DOI: 10.1038/s41431-022-01043-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/23/2021] [Accepted: 01/07/2022] [Indexed: 12/29/2022] Open
Abstract
Dilated cardiomyopathy (DCM) is characterized by cardiac enlargement and impaired ventricular contractility leading to heart failure. A single report identified variants in leiomodin-2 (LMOD2) as a cause of neonatally-lethal DCM. Here, we describe two siblings with DCM who died shortly after birth due to heart failure. Exome sequencing identified a homozygous LMOD2 variant in both siblings, (GRCh38)chr7:g.123656237G > A; NM_207163.2:c.273 + 1G > A, ablating the donor 5' splice-site of intron-1. Pre-mRNA splicing studies and western blot analysis on cDNA derived from proband cardiac tissue, MyoD-transduced proband skin fibroblasts and HEK293 cells transfected with LMOD2 gene constructs established variant-associated absence of canonically spliced LMOD2 mRNA and full-length LMOD2 protein. Immunostaining of proband heart tissue unveiled abnormally short actin-thin filaments. Our data are consistent with LMOD2 c.273 + 1G > A abolishing/reducing LMOD2 transcript expression by: (1) variant-associated perturbation in initiation of transcription due to ablation of the intron-1 donor; and/or (2) degradation of aberrant LMOD2 transcripts (resulting from use of alternative transcription start-sites or cryptic splice-sites) by nonsense-mediated decay. LMOD2 expression is critical for life and the absence of LMOD2 is associated with thin filament shortening and severe cardiac contractile dysfunction. This study describes the first splice-site variant in LMOD2 and confirms the role of LMOD2 variants in DCM.
Collapse
Affiliation(s)
- Michaela Yuen
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Westmead, NSW, Australia.
- Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Camperdown, NSW, Australia.
| | - Lisa Worgan
- Department of Medical Genomics, Royal Prince Alfred Hospital, Camperdown, NSW, Australia
| | - Jessika Iwanski
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Christopher T Pappas
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Himanshu Joshi
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Jared M Churko
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Susan Arbuckle
- Department of Histopathology, The Children's Hospital at Westmead, Westmead, NSW, Australia
| | - Edwin P Kirk
- New South Wales Health Pathology, Randwick Genomics Laboratory, Randwick, NSW, Australia
- School of Women's and Children's Health, University of New South Wales, Randwick, NSW, Australia
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia
| | - Ying Zhu
- New South Wales Health Pathology, Randwick Genomics Laboratory, Randwick, NSW, Australia
| | - Tony Roscioli
- New South Wales Health Pathology, Randwick Genomics Laboratory, Randwick, NSW, Australia
- Centre for Clinical Genetics, Sydney Children's Hospital, Randwick, NSW, Australia
- Neuroscience Research Australia (NeuRA), University of New South Wales, Sydney, NSW, Australia
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Sandra T Cooper
- Kids Neuroscience Centre, The Children's Hospital at Westmead, Westmead, NSW, Australia
- Discipline of Child and Adolescent Health, Sydney Medical School, University of Sydney, Camperdown, NSW, Australia
- The Children's Medical Research Institute, Westmead, NSW, Australia
| |
Collapse
|
30
|
Liu K, Lu L, Chen S, Gu B, Cai H, Wang Y, Cai W. Loss-of-function variants within LMOD1 actin-binding site 2 cause pediatric intestinal pseudo-obstruction by impairing protein stability and actin nucleation. FASEB J 2022; 36:e22194. [PMID: 35170814 DOI: 10.1096/fj.202101395r] [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: 09/02/2021] [Revised: 01/12/2022] [Accepted: 01/21/2022] [Indexed: 11/11/2022]
Abstract
The leiomodin1 (LMOD1) gene, encoding a potent actin nucleator, was recently reported as a potential pathogenic gene of megacystis-microcolon-intestinal hypoperistalsis syndrome (MMIHS, OMIM 619362). However, only a single patient has been reported to have LMOD1 mutations, and the underlying pathogenic mechanism remains unknown. Here, we described a male infant with LMOD1 mutations presenting typical symptoms of pediatric intestinal pseudo-obstruction (PIPO) but without megacystis and microcolon. Two compound heterozygous missense variants (c.1106C>T, p.T369M; c.1262G>A, p.R421H) were identified, both affecting highly conserved amino acid residues within the second actin-binding site (ABS2) domain of LMOD1. Expression analysis showed that both variants resulted in significantly reduced protein amounts, especially for p.T369M, which was almost undetectable. The reduction was only partially rescued by the proteasome inhibitor MG-132, indicating that there might be proteasome-independent pathways involved in the degradation of the mutant proteins. Molecular modeling showed that variant p.T369M impaired the local protein conformation of the ABS2 domain, while variant p.R421H directly impaired the intermolecular interaction between ABS2 and actin. Accordingly, both variants significantly damaged LMOD1-mediated actin nucleation. These findings provide further human genetic evidence supporting LMOD1 as a pathogenic gene underlying visceral myopathy including PIPO and MMIHS, strengthen the critical role of ABS2 domain in LMOD1-mediated actin nucleation, and moreover, reveal an unrecognized role of ABS2 in protein stability.
Collapse
Affiliation(s)
- Keqiang Liu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Lina Lu
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shanshan Chen
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Beilin Gu
- Shanghai Institute for Pediatric Research, Shanghai, China
| | - Hui Cai
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Wang
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China
| | - Wei Cai
- Division of Pediatric Gastroenterology and Nutrition, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Institute for Pediatric Research, Shanghai, China.,Shanghai Key Laboratory of Pediatric Gastroenterology and Nutrition, Shanghai, China.,Department of Pediatric Surgery, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
31
|
Berger J, Berger S, Mok YSG, Li M, Tarakci H, Currie PD. Genetic dissection of novel myopathy models reveals a role of CapZα and Leiomodin 3 during myofibril elongation. PLoS Genet 2022; 18:e1010066. [PMID: 35148320 PMCID: PMC8870547 DOI: 10.1371/journal.pgen.1010066] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 02/24/2022] [Accepted: 02/01/2022] [Indexed: 12/18/2022] Open
Abstract
Myofibrils within skeletal muscle are composed of sarcomeres that generate force by contraction when their myosin-rich thick filaments slide past actin-based thin filaments. Although mutations in components of the sarcomere are a major cause of human disease, the highly complex process of sarcomere assembly is not fully understood. Current models of thin filament assembly highlight a central role for filament capping proteins, which can be divided into three protein families, each ascribed with separate roles in thin filament assembly. CapZ proteins have been shown to bind the Z-disc protein α-actinin to form an anchoring complex for thin filaments and actin polymerisation. Subsequent thin filaments extension dynamics are thought to be facilitated by Leiomodins (Lmods) and thin filament assembly is concluded by Tropomodulins (Tmods) that specifically cap the pointed end of thin filaments. To study thin filament assembly in vivo, single and compound loss-of-function zebrafish mutants within distinct classes of capping proteins were analysed. The generated lmod3- and capza1b-deficient zebrafish exhibited aspects of the pathology caused by variations in their human orthologs. Although loss of the analysed main capping proteins of the skeletal muscle, capza1b, capza1a, lmod3 and tmod4, resulted in sarcomere defects, residual organised sarcomeres were formed within the assessed mutants, indicating that these proteins are not essential for the initial myofibril assembly. Furthermore, detected similarity and location of myofibril defects, apparent at the peripheral ends of myofibres of both Lmod3- and CapZα-deficient mutants, suggest a function in longitudinal myofibril growth for both proteins, which is molecularly distinct to the function of Tmod4. The force-generating contractile apparatus is a highly organised structure mainly composed of thick and thin filaments of uniform length. Three families of capping proteins are described to play a role in the regulation of thin filament length. Current models suggest that thin filament assembly is initiated by CapZ, extended by Leiomodins (Lmods) and concluded by Tropomodulins (Tmods). To better understand the role of these capping proteins, we analysed single and double loss-of-function zebrafish mutants for these capping proteins. We find that lmod3- and capza1b-deficient zebrafish model aspects of the human disorders caused by variations in their orthologs. Surprisingly, although pivotal for sarcomere formation, our results reveal that none of the analysed capping proteins, capza1b, capza1a, lmod3 and tmod4, are absolutely required for thin filament assembly, as suggested by current models. Our study further indicates that the roles of CapZ and Lmod3 are distinct from Tmod4. Both Lmod3- as well as CapZα-deficient mutants feature specific defects at the peripheral ends of muscle cells. We conclude that, in addition to their non-essential role during thin filament assembly, both Lmod3- and CapZα proteins may function in the longitudinal growth of the contractile apparatus.
Collapse
Affiliation(s)
- Joachim Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
- Victoria Node, EMBL Australia, Clayton, Australia
- * E-mail: (JB); (PDC)
| | - Silke Berger
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
- Victoria Node, EMBL Australia, Clayton, Australia
| | - Yu Shan G. Mok
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
- Victoria Node, EMBL Australia, Clayton, Australia
| | - Mei Li
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
- Victoria Node, EMBL Australia, Clayton, Australia
| | - Hakan Tarakci
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
- Victoria Node, EMBL Australia, Clayton, Australia
| | - Peter D. Currie
- Australian Regenerative Medicine Institute, Monash University, Clayton, Australia
- Victoria Node, EMBL Australia, Clayton, Australia
- * E-mail: (JB); (PDC)
| |
Collapse
|
32
|
Wenz E, Tafti M, Bassetti CL. LMOD3 gene variant in familial periodic hypersomnolence. Sleep Med 2022; 91:105-108. [DOI: 10.1016/j.sleep.2022.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/14/2022] [Accepted: 02/19/2022] [Indexed: 10/19/2022]
|
33
|
Kotecha UH, Mistri M, Rayabarapu P, Shah P, Shah N. The diagnostic utility of exome-based carrier screening in families with a positive family history. Am J Med Genet A 2022; 188:1323-1333. [PMID: 34997808 DOI: 10.1002/ajmg.a.62633] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 10/15/2021] [Accepted: 12/26/2021] [Indexed: 11/09/2022]
Abstract
Identification of disease-causing variants in families with a history of a suspected recessive disorder is essential for appropriate counseling and reproductive decision making. The present case series depicts the utility of whole exome-based phenotypes-driven carrier analysis in 14 families with a positive family history. A phenotype-based analysis revealed a putative diagnostic yield of 71.4%. Proband sample, though insufficient, was available in only one family, which allowed the diagnosis to be confirmed. In the remaining nine families, despite the detection of heterozygous pathogenic/likely pathogenic variants, only a putative diagnosis was possible due to incomplete proband phenotyping as well as nonavailability of proband samples. We describe the youngest known patient homozygous for a likely pathogenic variant in PPP1R21. He is currently asymptomatic at 7 days of life and has a simplified gyral pattern on neuroimaging. The case series, though small, captures the challenges in the diagnosis of genetic disorders in low to middle income countries with in-equitable health care access. It reinforces the significance of detailed phenotyping in the proband as well as the importance of DNA storage for a conclusive diagnosis. A recurring post-test counseling challenge was risk ascertainment and reproductive decision making in subsequent pregnancies if the detected pathogenic/likely pathogenic variants are co-inherited, in families with a putative diagnosis. When opted for, prenatal testing in such a scenario would be limited in its ability to comment on the fetal status with respect to the disorder in the proband.
Collapse
Affiliation(s)
| | - Mehul Mistri
- Neuberg Centre for Genomic Medicine, Ahmedabad, India
| | | | - Parth Shah
- Neuberg Centre for Genomic Medicine, Ahmedabad, India
| | - Nidhi Shah
- Neuberg Centre for Genomic Medicine, Ahmedabad, India
| |
Collapse
|
34
|
Hou X, Wang L, Zhao F, Liu X, Gao H, Shi L, Yan H, Wang L, Zhang L. Genome-Wide Expression Profiling of mRNAs, lncRNAs and circRNAs in Skeletal Muscle of Two Different Pig Breeds. Animals (Basel) 2021; 11:ani11113169. [PMID: 34827901 PMCID: PMC8614396 DOI: 10.3390/ani11113169] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/04/2021] [Accepted: 11/04/2021] [Indexed: 01/02/2023] Open
Abstract
Simple Summary Variation exists in muscle-related traits, such as muscle growth and meat quality, between obese and lean pigs. In this study, the transcriptome profiles of skeletal muscle between Beijing Blackand Yorkshire pigs were characterized to explore the molecular mechanism underlying skeletal muscle-relatedtraits. Gene Ontology (GO) and KEGG pathway enrichment analyses showed that differentially expressed mRNAs, lncRNAs, and circRNAs involved in skeletal muscle development and fatty acid metabolism played a key role in the determination of muscle-related traits between different pig breeds. These results provide candidate genes responsible for muscle phenotypic variation and are valuable for pig breeding. Abstract RNA-Seq technology is widely used to analyze global changes in the transcriptome and investigate the influence on relevant phenotypic traits. Beijing Black pigs show differences in growth rate and meat quality compared to western pig breeds. However, the molecular mechanisms responsible for such phenotypic differences remain unknown. In this study, longissimus dorsi muscles from Beijing Black and Yorkshire pigs were used to construct RNA libraries and perform RNA-seq. Significantly different expressions were observed in 1051 mRNAs, 322 lncRNAs, and 82 circRNAs. GO and KEGG pathway annotation showed that differentially expressed mRNAs participated in skeletal muscle development and fatty acid metabolism, which determined the muscle-related traits. To explore the regulatory role of lncRNAs, the cis and trans-target genes were predicted and these lncRNAswere involved in the biological processes related to skeletal muscle development and fatty acid metabolismvia their target genes. CircRNAs play a ceRNA role by binding to miRNAs. Therefore, the potential miRNAs of differentially expressed circRNAs were predicted and interaction networks among circRNAs, miRNAs, and key regulatory mRNAs were constructed to illustrate the function of circRNAs underlying skeletal muscle development and fatty acid metabolism. This study provides new clues for elucidating muscle phenotypic variation in pigs.
Collapse
|
35
|
Molecular and cellular basis of genetically inherited skeletal muscle disorders. Nat Rev Mol Cell Biol 2021; 22:713-732. [PMID: 34257452 PMCID: PMC9686310 DOI: 10.1038/s41580-021-00389-z] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/04/2021] [Indexed: 02/06/2023]
Abstract
Neuromuscular disorders comprise a diverse group of human inborn diseases that arise from defects in the structure and/or function of the muscle tissue - encompassing the muscle cells (myofibres) themselves and their extracellular matrix - or muscle fibre innervation. Since the identification in 1987 of the first genetic lesion associated with a neuromuscular disorder - mutations in dystrophin as an underlying cause of Duchenne muscular dystrophy - the field has made tremendous progress in understanding the genetic basis of these diseases, with pathogenic variants in more than 500 genes now identified as underlying causes of neuromuscular disorders. The subset of neuromuscular disorders that affect skeletal muscle are referred to as myopathies or muscular dystrophies, and are due to variants in genes encoding muscle proteins. Many of these proteins provide structural stability to the myofibres or function in regulating sarcolemmal integrity, whereas others are involved in protein turnover, intracellular trafficking, calcium handling and electrical excitability - processes that ensure myofibre resistance to stress and their primary activity in muscle contraction. In this Review, we discuss how defects in muscle proteins give rise to muscle dysfunction, and ultimately to disease, with a focus on pathologies that are most common, best understood and that provide the most insight into muscle biology.
Collapse
|
36
|
Laitila J, Wallgren-Pettersson C. Recent advances in nemaline myopathy. Neuromuscul Disord 2021; 31:955-967. [PMID: 34561123 DOI: 10.1016/j.nmd.2021.07.012] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Revised: 07/14/2021] [Accepted: 07/16/2021] [Indexed: 11/18/2022]
Abstract
The nemaline myopathies constitute a large proportion of the congenital or structural myopathies. Common to all patients is muscle weakness and the presence in the muscle biopsy of nemaline rods. The causative genes are at least twelve, encoding structural or regulatory proteins of the thin filament, and the clinical picture as well as the histological appearance on muscle biopsy vary widely. Here, we suggest a renewed clinical classification to replace the original one, summarise what is known about the pathogenesis from mutations in each causative gene to the forms of nemaline myopathy described to date, and provide perspectives on pathogenetic mechanisms possibly open to therapeutic modalities.
Collapse
Affiliation(s)
- Jenni Laitila
- The Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland; Department of Medical Genetics, Medicum, University of Helsinki, Finland; Department of Biomedical Sciences, University of Copenhagen, Denmark.
| | - Carina Wallgren-Pettersson
- The Folkhälsan Institute of Genetics, Folkhälsan Research Center, Helsinki, Finland; Department of Medical Genetics, Medicum, University of Helsinki, Finland
| |
Collapse
|
37
|
Whole-genome association study searching for QTL for Aeromonas salmonicida resistance in rainbow trout. Sci Rep 2021; 11:17857. [PMID: 34497310 PMCID: PMC8426485 DOI: 10.1038/s41598-021-97437-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
Aeromonas salmonicida subsp. salmonicida, the causative agent of furunculosis, has extensive negative effects on wild and farmed salmonids worldwide. Vaccination induces some protection under certain conditions but disease outbreaks occur even in vaccinated fish. Therefore, alternative disease control approaches are required to ensure the sustainable expansion of rainbow trout aquaculture. Selective breeding can be applied to enhance host resistance to pathogens. The present work used genome-wide association study (GWAS) to identify quantitative trait loci (QTL) associated with A. salmonicida resistance in rainbow trout. A total 798 rainbow trout exposed to A. salmonicida by bath challenge revealed 614 susceptible and 138 resistant fish. Genotyping was conducted using the 57 K single nucleotide polymorphism (SNP) array and the GWAS was performed for survival and time to death phenotypes. We identified a QTL on chromosome 16 and located positional candidate genes in the proximity of the most significant SNPs. In addition, samples from exposed fish were examined for expression of 24 immune-relevant genes indicating a systematic immune response to the infection. The present work demonstrated that resistance to A. salmonicida is moderately heritable with oligogenic architecture. These result will be useful for the future breeding programs for improving the natural resistance of rainbow trout against furunculosis.
Collapse
|
38
|
Jirka C, Pak JH, Grosgogeat CA, Marchetii MM, Gupta VA. Dysregulation of NRAP degradation by KLHL41 contributes to pathophysiology in nemaline myopathy. Hum Mol Genet 2021; 28:2549-2560. [PMID: 30986853 DOI: 10.1093/hmg/ddz078] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 03/29/2019] [Accepted: 04/08/2019] [Indexed: 12/13/2022] Open
Abstract
Nemaline myopathy (NM) is the most common form of congenital myopathy that results in hypotonia and muscle weakness. This disease is clinically and genetically heterogeneous, but three recently discovered genes in NM encode for members of the Kelch family of proteins. Kelch proteins act as substrate-specific adaptors for Cullin 3 (CUL3) E3 ubiquitin ligase to regulate protein turnover through the ubiquitin-proteasome machinery. Defects in thin filament formation and/or stability are key molecular processes that underlie the disease pathology in NM; however, the role of Kelch proteins in these processes in normal and diseases conditions remains elusive. Here, we describe a role of NM causing Kelch protein, KLHL41, in premyofibil-myofibil transition during skeletal muscle development through a regulation of the thin filament chaperone, nebulin-related anchoring protein (NRAP). KLHL41 binds to the thin filament chaperone NRAP and promotes ubiquitination and subsequent degradation of NRAP, a process that is critical for the formation of mature myofibrils. KLHL41 deficiency results in abnormal accumulation of NRAP in muscle cells. NRAP overexpression in transgenic zebrafish resulted in a severe myopathic phenotype and absence of mature myofibrils demonstrating a role in disease pathology. Reducing Nrap levels in KLHL41 deficient zebrafish rescues the structural and function defects associated with disease pathology. We conclude that defects in KLHL41-mediated ubiquitination of sarcomeric proteins contribute to structural and functional deficits in skeletal muscle. These findings further our understanding of how the sarcomere assembly is regulated by disease-causing factors in vivo, which will be imperative for developing mechanism-based specific therapeutic interventions.
Collapse
Affiliation(s)
- Caroline Jirka
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jasmine H Pak
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Claire A Grosgogeat
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Vandana A Gupta
- Division of Genetics, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
39
|
Vignier N, Chatzifrangkeskou M, Pinton L, Wioland H, Marais T, Lemaitre M, Le Dour C, Peccate C, Cardoso D, Schmitt A, Wu W, Biferi MG, Naouar N, Macquart C, Beuvin M, Decostre V, Bonne G, Romet-Lemonne G, Worman HJ, Tedesco FS, Jégou A, Muchir A. The non-muscle ADF/cofilin-1 controls sarcomeric actin filament integrity and force production in striated muscle laminopathies. Cell Rep 2021; 36:109601. [PMID: 34433058 PMCID: PMC8411111 DOI: 10.1016/j.celrep.2021.109601] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2020] [Revised: 06/09/2021] [Accepted: 08/04/2021] [Indexed: 12/11/2022] Open
Abstract
Cofilins are important for the regulation of the actin cytoskeleton, sarcomere organization, and force production. The role of cofilin-1, the non-muscle-specific isoform, in muscle function remains unclear. Mutations in LMNA encoding A-type lamins, intermediate filament proteins of the nuclear envelope, cause autosomal Emery-Dreifuss muscular dystrophy (EDMD). Here, we report increased cofilin-1 expression in LMNA mutant muscle cells caused by the inability of proteasome degradation, suggesting a protective role by ERK1/2. It is known that phosphorylated ERK1/2 directly binds to and catalyzes phosphorylation of the actin-depolymerizing factor cofilin-1 on Thr25. In vivo ectopic expression of cofilin-1, as well as its phosphorylated form on Thr25, impairs sarcomere structure and force generation. These findings present a mechanism that provides insight into the molecular pathogenesis of muscular dystrophies caused by LMNA mutations.
Collapse
Affiliation(s)
- Nicolas Vignier
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Maria Chatzifrangkeskou
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Luca Pinton
- Department of Cell and Developmental Biology, University College London, London, UK; Randall Centre for Cell and Molecular Biophysics, King's College London, London, UK
| | - Hugo Wioland
- Université de Paris, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Thibaut Marais
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Mégane Lemaitre
- Sorbonne Université, UMS28, Phénotypage du Petit Animal, Paris, France
| | - Caroline Le Dour
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Cécile Peccate
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Déborah Cardoso
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Alain Schmitt
- Université de Paris, INSERM, CNRS, Institut Cochin, 75005 Paris, France
| | - Wei Wu
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Maria-Grazia Biferi
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Naïra Naouar
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Coline Macquart
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Maud Beuvin
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Valérie Decostre
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | - Gisèle Bonne
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France
| | | | - Howard J Worman
- Department of Medicine, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA; Department of Pathology and Cell Biology, Vagelos College of Physicians and Surgeons, Columbia University, New York, NY, USA
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, London, UK; Dubowitz Neuromuscular Centre, UCL Great Ormond Street Institute of Child Health and Great Ormond Street Hospital for Children, London, UK; The Francis Crick Institute, London, UK
| | - Antoine Jégou
- Université de Paris, CNRS, Institut Jacques Monod, 75013 Paris, France
| | - Antoine Muchir
- Sorbonne Université, INSERM, Institut de Myologie, Centre de Recherche en Myologie, 75013 Paris, France.
| |
Collapse
|
40
|
Iwanski J, Gregorio CC, Colpan M. Redefining actin dynamics of the pointed-end complex in striated muscle. Trends Cell Biol 2021; 31:708-711. [PMID: 34266732 DOI: 10.1016/j.tcb.2021.06.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/14/2021] [Accepted: 06/16/2021] [Indexed: 11/15/2022]
Abstract
Striated muscle is intricately designed to provide efficient and powerful muscle contractions. Recently, a long sought-after missing component of the thin filament pointed-end machinery was discovered: cyclase-associated protein 2 (CAP2). CAP2 was identified as a crucial contributor to actin polymerization, striated muscle development, and severe muscle disease when mutated.
Collapse
Affiliation(s)
- Jessika Iwanski
- Department of Cellular and Molecular Medicine, and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ 85724, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine, and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ 85724, USA.
| | - Mert Colpan
- Department of Cellular and Molecular Medicine, and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ 85724, USA
| |
Collapse
|
41
|
de Winter JM, Gineste C, Minardi E, Brocca L, Rossi M, Borsboom T, Beggs AH, Bernard M, Bendahan D, Hwee DT, Malik FI, Pellegrino MA, Bottinelli R, Gondin J, Ottenheijm CAC. Acute and chronic tirasemtiv treatment improves in vivo and in vitro muscle performance in actin-based nemaline myopathy mice. Hum Mol Genet 2021; 30:1305-1320. [PMID: 33909041 PMCID: PMC8255131 DOI: 10.1093/hmg/ddab112] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/09/2021] [Accepted: 04/07/2021] [Indexed: 12/13/2022] Open
Abstract
Nemaline myopathy, a disease of the actin-based thin filament, is one of the most frequent congenital myopathies. To date, no specific therapy is available to treat muscle weakness in nemaline myopathy. We tested the ability of tirasemtiv, a fast skeletal troponin activator that targets the thin filament, to augment muscle force-both in vivo and in vitro-in a nemaline myopathy mouse model with a mutation (H40Y) in Acta1. In Acta1H40Y mice, treatment with tirasemtiv increased the force response of muscles to submaximal stimulation frequencies. This resulted in a reduced energetic cost of force generation, which increases the force production during a fatigue protocol. The inotropic effects of tirasemtiv were present in locomotor muscles and, albeit to a lesser extent, in respiratory muscles, and they persisted during chronic treatment, an important finding as respiratory failure is the main cause of death in patients with congenital myopathy. Finally, translational studies on permeabilized muscle fibers isolated from a biopsy of a patient with the ACTA1H40Y mutation revealed that at physiological Ca2+ concentrations, tirasemtiv increased force generation to values that were close to those generated in muscle fibers of healthy subjects. These findings indicate the therapeutic potential of fast skeletal muscle troponin activators to improve muscle function in nemaline myopathy due to the ACTA1H40Y mutation, and future studies should assess their merit for other forms of nemaline myopathy and for other congenital myopathies.
Collapse
Affiliation(s)
- Josine M de Winter
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam 1081 HV, The Netherlands
| | | | - Elisa Minardi
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Lorenza Brocca
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Maira Rossi
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
| | - Tamara Borsboom
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam 1081 HV, The Netherlands
| | - Alan H Beggs
- Division of Genetics and Genomics, The Manton Center for Orphan Disease Research, Boston Children's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Monique Bernard
- Aix-Marseille Univ, CNRS, CRMBM, UMR 7339, 13005 Marseille, France
| | - David Bendahan
- Aix-Marseille Univ, CNRS, CRMBM, UMR 7339, 13005 Marseille, France
| | - Darren T Hwee
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Fady I Malik
- Research and Early Development, Cytokinetics Inc., South San Francisco, CA 94080, USA
| | - Maria Antonietta Pellegrino
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
- Interdipartimental Centre for Biology and Sport Medicine, University of Pavia, Pavia 27100, Italy
| | - Roberto Bottinelli
- Department of Molecular Medicine, University of Pavia, Pavia 27100, Italy
- Istituti Clinici Maugeri (IRCCS), Scientific Institute of Pavia, Pavia 27100, Italy
| | - Julien Gondin
- Aix-Marseille Univ, CNRS, CRMBM, UMR 7339, 13005 Marseille, France
- Institut NeuroMyoGène, Université Claude Bernard Lyon 1, CNRS 5310, INSERM U1217, 69008, Lyon, France
| | - Coen A C Ottenheijm
- Department of Physiology, Amsterdam UMC (location VUmc), Amsterdam 1081 HV, The Netherlands
| |
Collapse
|
42
|
Laquerriere A, Jaber D, Abiusi E, Maluenda J, Mejlachowicz D, Vivanti A, Dieterich K, Stoeva R, Quevarec L, Nolent F, Biancalana V, Latour P, Sternberg D, Capri Y, Verloes A, Bessieres B, Loeuillet L, Attie-Bitach T, Martinovic J, Blesson S, Petit F, Beneteau C, Whalen S, Marguet F, Bouligand J, Héron D, Viot G, Amiel J, Amram D, Bellesme C, Bucourt M, Faivre L, Jouk PS, Khung S, Sigaudy S, Delezoide AL, Goldenberg A, Jacquemont ML, Lambert L, Layet V, Lyonnet S, Munnich A, Van Maldergem L, Piard J, Guimiot F, Landrieu P, Letard P, Pelluard F, Perrin L, Saint-Frison MH, Topaloglu H, Trestard L, Vincent-Delorme C, Amthor H, Barnerias C, Benachi A, Bieth E, Boucher E, Cormier-Daire V, Delahaye-Duriez A, Desguerre I, Eymard B, Francannet C, Grotto S, Lacombe D, Laffargue F, Legendre M, Martin-Coignard D, Mégarbané A, Mercier S, Nizon M, Rigonnot L, Prieur F, Quélin C, Ranjatoelina-Randrianaivo H, Resta N, Toutain A, Verhelst H, Vincent M, Colin E, Fallet-Bianco C, Granier M, Grigorescu R, Saada J, Gonzales M, Guiochon-Mantel A, Bessereau JL, Tawk M, Gut I, Gitiaux C, Melki J. Phenotypic spectrum and genomics of undiagnosed arthrogryposis multiplex congenita. J Med Genet 2021; 59:559-567. [PMID: 33820833 PMCID: PMC9132874 DOI: 10.1136/jmedgenet-2020-107595] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/23/2021] [Accepted: 03/14/2021] [Indexed: 12/16/2022]
Abstract
BACKGROUND Arthrogryposis multiplex congenita (AMC) is characterised by congenital joint contractures in two or more body areas. AMC exhibits wide phenotypic and genetic heterogeneity. Our goals were to improve the genetic diagnosis rates of AMC, to evaluate the added value of whole exome sequencing (WES) compared with targeted exome sequencing (TES) and to identify new genes in 315 unrelated undiagnosed AMC families. METHODS Several genomic approaches were used including genetic mapping of disease loci in multiplex or consanguineous families, TES then WES. Sanger sequencing was performed to identify or validate variants. RESULTS We achieved disease gene identification in 52.7% of AMC index patients including nine recently identified genes (CNTNAP1, MAGEL2, ADGRG6, ADCY6, GLDN, LGI4, LMOD3, UNC50 and SCN1A). Moreover, we identified pathogenic variants in ASXL3 and STAC3 expanding the phenotypes associated with these genes. The most frequent cause of AMC was a primary involvement of skeletal muscle (40%) followed by brain (22%). The most frequent mode of inheritance is autosomal recessive (66.3% of patients). In sporadic patients born to non-consanguineous parents (n=60), de novo dominant autosomal or X linked variants were observed in 30 of them (50%). CONCLUSION New genes recently identified in AMC represent 21% of causing genes in our cohort. A high proportion of de novo variants were observed indicating that this mechanism plays a prominent part in this developmental disease. Our data showed the added value of WES when compared with TES due to the larger clinical spectrum of some disease genes than initially described and the identification of novel genes.
Collapse
Affiliation(s)
- Annie Laquerriere
- Normandie Univ, UNIROUEN, INSERM U1245; Rouen University Hospital, Department of Pathology, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Dana Jaber
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France
| | - Emanuela Abiusi
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France.,Fondazione Policlinico Universitario A. Gemelli Istituto di Ricovero e Cura a Carattere Scientifico and Sezione di Medicina Genomica, Dipartimento di Scienze della Vita e Sanità Pubblica, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Jérome Maluenda
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France
| | - Dan Mejlachowicz
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France
| | - Alexandre Vivanti
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France
| | - Klaus Dieterich
- Univ. Grenoble Alpes, Inserm, U1209, CHU Grenoble Alpes, Grenoble, France
| | - Radka Stoeva
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France.,Department of Medical Genetics, Le Mans Hospital, Le Mans, France
| | - Loic Quevarec
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France
| | - Flora Nolent
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France
| | - Valerie Biancalana
- Laboratoire Diagnostic Génétique, CHRU, Strasbourg; Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), INSERM U964, CNRS UMR 7104, Fédération de Médecine Translationnelle de Strasbourg, Université de Strasbourg, Illkirch, France
| | - Philippe Latour
- Centre de Biologie Est, Hospices Civils de Lyon, Bron, France
| | - Damien Sternberg
- Service de Biochimie Métabolique et Centre de Génétique, APHP. Sorbonne Université, GH Pitié-Salpêtrière; Centre of Research in Myology, Sorbonne University, UMRS 974, Paris, France
| | - Yline Capri
- Département de Génétique, Assistance publique-Hopitaux de Paris (AP-HP), Hopital Robert Debré, Paris, France
| | - Alain Verloes
- Département de Génétique, Assistance publique-Hopitaux de Paris (AP-HP), Hopital Robert Debré, Paris, France
| | - Bettina Bessieres
- Unité d'Embryofoetopathologie, Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Laurence Loeuillet
- Unité d'Embryofoetopathologie, Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Tania Attie-Bitach
- Unité d'Embryofoetopathologie, Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Jelena Martinovic
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France.,Unité d'Embryofoetopathologie, Hôpital Antoine Béclère, APHP, Clamart, France
| | - Sophie Blesson
- Service de Génétique, Unité de Génétique Clinique, CHRU de Tours, Hôpital Bretonneau, Tours, France
| | - Florence Petit
- Service de Génétique Clinique Guy Fontaine, CHU Lille, Lille, France
| | - Claire Beneteau
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Nantes; Institut du Thorax, INSERM, CNRS, Université de Nantes, Nantes, France
| | - Sandra Whalen
- UF de Génétique clinique et Centre de Référence Maladies Rares des Anomalies du Développement et Syndromes Malformatifs, APHP. Sorbonne Université, Hôpital Armand Trousseau, Paris, France
| | - Florent Marguet
- Normandie Univ, UNIROUEN, INSERM U1245; Rouen University Hospital, Department of Pathology, Normandy Centre for Genomic and Personalized Medicine, Rouen, France
| | - Jerome Bouligand
- Laboratoire de Génétique moléculaire, Pharmacogénétique et Hormonologie, Hôpital Bicêtre, APHP Université Paris Saclay, Le Kremlin-Bicêtre; Inserm UMR_S 1185, Faculté de médecine Paris Saclay, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Delphine Héron
- Département de Génétique, APHP Sorbonne Université, Hôpital Pitié-Salpêtrière et Trousseau, PARIS, France
| | - Géraldine Viot
- Unité de Génétique, Clinique de la Muette, Paris, France
| | - Jeanne Amiel
- Service de Génétique Clinique, Centre de référence pour les maladies osseuses constitutionnelles APHP, Hôpital Necker-Enfants Malades; Université de Paris, UMR1163, INSERM, Institut Imagine, Paris, France
| | - Daniel Amram
- Unité de Génétique Clinique, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Céline Bellesme
- Department of Pediatric Neurology, APHP-Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Martine Bucourt
- Service d'Histologie, Embryologie, et Cytogénétique, Hôpital Jean Verdier, APHP, Bondy, France
| | - Laurence Faivre
- Centre de Génétique et Centre de référence Anomalies du Développement et Syndromes Malformatifs, FHU TRANSLAD, Hôpital d'Enfants, CHU Dijon; UMR-Inserm 1231 GAD team, Génétique des Anomalies du développement, Université de Bourgogne Franche-Comté, Dijon, France
| | - Pierre-Simon Jouk
- Univ. Grenoble Alpes, Inserm, U1209, CHU Grenoble Alpes, Grenoble, France
| | - Suonavy Khung
- Unité Fonctionnelle de Fœtopathologie, Hôpital Universitaire Robert Debré; Inserm UMR 1141, Paris, France
| | - Sabine Sigaudy
- Département de Génétique Médicale, Hôpital Timone Enfant, Marseille, France
| | - Anne-Lise Delezoide
- Unité Fonctionnelle de Fœtopathologie, Hôpital Universitaire Robert Debré; Inserm UMR 1141, Paris, France
| | - Alice Goldenberg
- Department of Genetics and Reference Center for Developmental Disorders, Normandy Center for Genomic and Personalized Medicine, Normandie Univ, UNIROUEN, Inserm U1245 and Rouen University Hospital, Rouen, France
| | - Marie-Line Jacquemont
- UF de Génétique Médicale, CHU la Réunion, site GHSR, Ile de La Réunion, Saint-Pierre, France
| | | | - Valérie Layet
- Consultations de Génétique, Groupe Hospitalier du Havre, Le Havre, France
| | - Stanislas Lyonnet
- Imagine Institute, INSERM UMR 1163, Université de Paris; Fédération de Génétique Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Paris, France
| | - Arnold Munnich
- Imagine Institute, INSERM UMR 1163, Université de Paris; Fédération de Génétique Médicale, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Paris, France
| | | | - Juliette Piard
- Centre de Génétique Humaine, Université de Franche-Comté, Besançon, France
| | - Fabien Guimiot
- Unité Fonctionnelle de Fœtopathologie, Hôpital Universitaire Robert Debré; Inserm UMR 1141, Paris, France
| | - Pierre Landrieu
- Department of Pediatric Neurology, APHP-Bicêtre Hospital, Le Kremlin-Bicêtre, France
| | - Pascaline Letard
- Service d'Histologie, Embryologie, et Cytogénétique, Hôpital Jean Verdier, APHP, Bondy, France
| | - Fanny Pelluard
- UMR U1053, INSERM et Université de Bordeaux; Unité de fœtopathologie, Service de pathologie, CHU de Bordeaux, Bordeaux, France
| | - Laurence Perrin
- Département de Génétique, Assistance publique-Hopitaux de Paris (AP-HP), Hopital Robert Debré, Paris, France
| | - Marie-Hélène Saint-Frison
- Unité Fonctionnelle de Fœtopathologie, Hôpital Universitaire Robert Debré; Inserm UMR 1141, Paris, France
| | - Haluk Topaloglu
- Yeditepe University Deparment of Pediatrics, Istanbul, Turkey
| | | | | | - Helge Amthor
- Neuromuscular Reference Centre, Pediatric Department, University Hospital Raymond Poincaré, Garches, France
| | - Christine Barnerias
- Service de Neuropédiatrie, CR Neuromusculaire Necker, Hôpital Necker- Enfants Malades, Paris, France
| | - Alexandra Benachi
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France.,Service de Gynécologie-Obstétrique, Hôpital Antoine Béclère, AP-HP, Clamart, France
| | - Eric Bieth
- Service de Génétique Médicale, Hopital Purpan, Toulouse, France
| | - Elise Boucher
- Centre de Génétique Humaine, Université de Franche-Comté, Besançon, France
| | - Valerie Cormier-Daire
- Service de Génétique Clinique, Centre de référence pour les maladies osseuses constitutionnelles APHP, Hôpital Necker-Enfants Malades; Université de Paris, UMR1163, INSERM, Institut Imagine, Paris, France
| | - Andrée Delahaye-Duriez
- Service d'Histologie, Embryologie, et Cytogénétique, Hôpital Jean Verdier, APHP, Bondy, France.,Université de Paris, NeuroDiderot, Inserm, Paris, France
| | - Isabelle Desguerre
- Service de Neuropédiatrie, CR Neuromusculaire Necker, Hôpital Necker- Enfants Malades, Paris, France
| | - Bruno Eymard
- Sorbonne Université, GH Pitié-Salpêtrière, Paris, France
| | - Christine Francannet
- Service de génétique médicale et centre de référence des anomalies du développement et des déficits intellectuels rares, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - Sarah Grotto
- Maternité Port-Royal, AP-HP, Hôpital Cochin, Paris, France
| | - Didier Lacombe
- Service de Génétique Médicale, CHU Bordeaux, Hopital Pellegrin, Bordeaux, France
| | - Fanny Laffargue
- Service de génétique médicale et centre de référence des anomalies du développement et des déficits intellectuels rares, CHU de Clermont-Ferrand, Clermont-Ferrand, France
| | - Marine Legendre
- Service de Génétique Médicale, CHU Bordeaux, Hopital Pellegrin, Bordeaux, France
| | | | - André Mégarbané
- Department of Human Genetics, Gilbert and Rose-Marie Ghagoury School of Medicine, Lebanese American University, Byblos, Lebanon
| | - Sandra Mercier
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Nantes; Institut du Thorax, INSERM, CNRS, Université de Nantes, Nantes, France
| | - Mathilde Nizon
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Nantes; Institut du Thorax, INSERM, CNRS, Université de Nantes, Nantes, France
| | - Luc Rigonnot
- Service de gynécologie obstétrique, Centre Hospitalier Sud Francilien, Corbeil Essonnes, France
| | - Fabienne Prieur
- Service de Génétique Clinique, CHU de Saint Etienne, Saint-Etienne, France
| | - Chloé Quélin
- Service de Génétique Clinique, CLAD Ouest, CHU Rennes, F-35033 RENNES, France
| | | | - Nicoletta Resta
- Department of Biomedical Sciences and Human Oncology (DIMO), Medical Genetics, University of Bari "Aldo Moro", Bari, Italy
| | - Annick Toutain
- Service de Génétique, Centre Hospitalier Universitaire de Tours; UMR 1253, iBrain, Université de Tours, Inserm, Tours, France
| | - Helene Verhelst
- Department of Pediatrics, Division of Pediatric Neurology, Ghent University Hospital, Ghent, Belgium
| | - Marie Vincent
- Service de Génétique Médicale, Centre Hospitalier Universitaire de Nantes; Institut du Thorax, INSERM, CNRS, Université de Nantes, Nantes, France
| | - Estelle Colin
- Service de Génétique Médicale, CHU d'Angers, Angers, France
| | | | - Michèle Granier
- Neonatology and Neonatal Intensive Care Unit, Centre Hospitalier Sud Francilien, Corbeil Essonnes, France
| | - Romulus Grigorescu
- Unité de Génétique du Développement fœtal, Département de Génétique et Embryologie médicales, CHU Paris Est, Hôpital d'Enfants Armand-Trousseau, Paris, France
| | - Julien Saada
- Service de Gynécologie-Obstétrique, Hôpital Antoine Béclère, AP-HP, Clamart, France
| | - Marie Gonzales
- Unité d'Embryofoetopathologie, Service d'Histologie-Embryologie-Cytogénétique, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Anne Guiochon-Mantel
- Laboratoire de Génétique moléculaire, Pharmacogénétique et Hormonologie, Hôpital Bicêtre, APHP Université Paris Saclay, Le Kremlin-Bicêtre; Inserm UMR_S 1185, Faculté de médecine Paris Saclay, Université Paris Saclay, Le Kremlin-Bicêtre, France
| | - Jean-Louis Bessereau
- Univ Lyon, Université Claude Bernard Lyon 1, CNRS UMR 5310, INSERM U 1217, Institut NeuroMyoGène, Lyon, France
| | - Marcel Tawk
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France
| | - Ivo Gut
- CNAG-CRG, Centre for Genomic Regulation (CRG), Barcelona Institute of Science and Technology (BIST); Universitat Pompeu Fabra (UPF), Barcelona, Spain
| | - Cyril Gitiaux
- Unité de Neurophysiologie Clinique, Centre de référence des maladies neuromusculaires, Hôpital Necker Enfants Malades, APHP, Université de Paris, Paris, France
| | - Judith Melki
- Institut National de la Santé et de la Recherche Médicale (Inserm), UMR-1195, Université Paris Saclay, Le Kremlin-Bicetre, France .,Unité de Génétique Médicale, Centre de référence des anomalies du développement et syndromes malformatifs d'Île-de-France, APHP, Le Kremlin Bicêtre, France
| |
Collapse
|
43
|
Colpan M, Iwanski J, Gregorio CC. CAP2 is a regulator of actin pointed end dynamics and myofibrillogenesis in cardiac muscle. Commun Biol 2021; 4:365. [PMID: 33742108 PMCID: PMC7979805 DOI: 10.1038/s42003-021-01893-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 02/22/2021] [Indexed: 01/31/2023] Open
Abstract
The precise assembly of actin-based thin filaments is crucial for muscle contraction. Dysregulation of actin dynamics at thin filament pointed ends results in skeletal and cardiac myopathies. Here, we discovered adenylyl cyclase-associated protein 2 (CAP2) as a unique component of thin filament pointed ends in cardiac muscle. CAP2 has critical functions in cardiomyocytes as it depolymerizes and inhibits actin incorporation into thin filaments. Strikingly distinct from other pointed-end proteins, CAP2's function is not enhanced but inhibited by tropomyosin and it does not directly control thin filament lengths. Furthermore, CAP2 plays an essential role in cardiomyocyte maturation by modulating pre-sarcomeric actin assembly and regulating α-actin composition in mature thin filaments. Identification of CAP2's multifunctional roles provides missing links in our understanding of how thin filament architecture is regulated in striated muscle and it reveals there are additional factors, beyond Tmod1 and Lmod2, that modulate actin dynamics at thin filament pointed ends.
Collapse
Affiliation(s)
- Mert Colpan
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Jessika Iwanski
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA
| | - Carol C Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
44
|
Evangelista T, Lornage X, Carlier PG, Bassez G, Brochier G, Chanut A, Lacène E, Bui MT, Metay C, Oppermann U, Böhm J, Laporte J, Romero NB. A Heterozygous Mutation in the Filamin C Gene Causes an Unusual Nemaline Myopathy With Ring Fibers. J Neuropathol Exp Neurol 2021; 79:908-914. [PMID: 32607581 DOI: 10.1093/jnen/nlaa052] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Indexed: 11/13/2022] Open
Abstract
Autosomal dominant pathogenic variants in the filamin C gene (FLNC) have been associated with myofibrillar myopathies, distal myopathies, and isolated cardiomyopathies. Mutations in different functional domains of FLNC can cause various clinical phenotypes. A novel heterozygous missense variant c.608G>A, p.(Cys203Tyr) in the actin binding domain of FLCN was found to cause an upper limb distal myopathy (MIM #614065). The muscle MRI findings are similar to those observed in FLNC-myofibrillar myopathy (MIM #609524). However, the muscle biopsy revealed >20% of muscle fibers with nemaline bodies, in addition to numerous ring fibers and a predominance of type 1 fibers. Overall, this case shows some unique and rare aspects of FLNC-myopathy constituting a new morphologic phenotype of FLNC-related myopathies.
Collapse
Affiliation(s)
- Teresinha Evangelista
- From the Neuromuscular Morphology Unit, Myology Institute.,Sorbonne Université, AP-HP, INSERM, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, Groupe Hospitalier Universitaire La Pitié-Salpêtrière, Paris, France
| | - Xavière Lornage
- Department of Translational Medicine and Neurogenetics, IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | | | - Guillaume Bassez
- From the Neuromuscular Morphology Unit, Myology Institute.,Sorbonne Université, AP-HP, INSERM, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, Groupe Hospitalier Universitaire La Pitié-Salpêtrière, Paris, France
| | - Guy Brochier
- From the Neuromuscular Morphology Unit, Myology Institute.,Sorbonne Université, AP-HP, INSERM, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, Groupe Hospitalier Universitaire La Pitié-Salpêtrière, Paris, France
| | - Anais Chanut
- From the Neuromuscular Morphology Unit, Myology Institute
| | - Emmanuelle Lacène
- From the Neuromuscular Morphology Unit, Myology Institute.,Sorbonne Université, AP-HP, INSERM, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, Groupe Hospitalier Universitaire La Pitié-Salpêtrière, Paris, France
| | - Mai-Thao Bui
- From the Neuromuscular Morphology Unit, Myology Institute
| | - Corinne Metay
- AP-HP, Centre de Génétique Moléculaire et Chromosomique, UF de Cardiomyogénétique et Myogénétique Moléculaire et Cellulaire, Groupe Hospitalier Universitaire La Pitié-Salpêtrière, Paris, France
| | - Ursula Oppermann
- Department of Translational Medicine and Neurogenetics, IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Johann Böhm
- Department of Translational Medicine and Neurogenetics, IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Jocelyn Laporte
- Department of Translational Medicine and Neurogenetics, IGBMC (Institut de Génétique et de Biologie Moléculaire et Cellulaire), INSERM U1258, CNRS UMR7104, Université de Strasbourg, Illkirch, France
| | - Norma B Romero
- From the Neuromuscular Morphology Unit, Myology Institute.,Sorbonne Université, AP-HP, INSERM, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, Groupe Hospitalier Universitaire La Pitié-Salpêtrière, Paris, France
| |
Collapse
|
45
|
Amburgey K, Acker M, Saeed S, Amin R, Beggs AH, Bönnemann CG, Brudno M, Constantinescu A, Dastgir J, Diallo M, Genetti CA, Glueck M, Hewson S, Hum C, Jain MS, Lawlor MW, Meyer OH, Nelson L, Sultanum N, Syed F, Tran T, Wang CH, Dowling JJ. A Cross-Sectional Study of Nemaline Myopathy. Neurology 2021; 96:e1425-e1436. [PMID: 33397769 PMCID: PMC8055318 DOI: 10.1212/wnl.0000000000011458] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2020] [Accepted: 12/07/2020] [Indexed: 02/05/2023] Open
Abstract
Objective Nemaline myopathy (NM) is a rare neuromuscular condition with clinical and genetic heterogeneity. To establish disease natural history, we performed a cross-sectional study of NM, complemented by longitudinal assessment and exploration of pilot outcome measures. Methods Fifty-seven individuals with NM were recruited at 2 family workshops, including 16 examined at both time points. Participants were evaluated by clinical history and physical examination. Functional outcome measures included the Motor Function Measure (MFM), pulmonary function tests (PFTs), myometry, goniometry, and bulbar assessments. Results The most common clinical classification was typical congenital (54%), whereas 42% had more severe presentations. Fifty-eight percent of individuals needed mechanical support, with 26% requiring wheelchair, tracheostomy, and feeding tube. The MFM scale was performed in 44 of 57 participants and showed reduced scores in most with little floor/ceiling effect. Of the 27 individuals completing PFTs, abnormal values were observed in 65%. Last, bulbar function was abnormal in all patients examined, as determined with a novel outcome measure. Genotypes included mutations in ACTA1 (18), NEB (20), and TPM2 (2). Seventeen individuals were genetically unresolved. Patients with pathogenic ACTA1 and NEB variants were largely similar in clinical phenotype. Patients without genetic resolution had more severe disease. Conclusion We present a comprehensive cross-sectional study of NM. Our data identify significant disabilities and support a relatively stable disease course. We identify a need for further diagnostic investigation for the genetically unresolved group. MFM, PFTs, and the slurp test were identified as promising outcome measures for future clinical trials.
Collapse
Affiliation(s)
- Kimberly Amburgey
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Meryl Acker
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Samia Saeed
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Reshma Amin
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Alan H Beggs
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Carsten G Bönnemann
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Michael Brudno
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Andrei Constantinescu
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Jahannaz Dastgir
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Mamadou Diallo
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Casie A Genetti
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Michael Glueck
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Stacy Hewson
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Courtney Hum
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Minal S Jain
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Michael W Lawlor
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Oscar H Meyer
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Leslie Nelson
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Nicole Sultanum
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Faiza Syed
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Tuyen Tran
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - Ching H Wang
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi
| | - James J Dowling
- From the Division of Neurology (K.A.), Genetics and Genome Biology (K.A., M.A., J.J.D., M.B., N.S.), Division of Respiratory Medicine (R.A., F.S., T.T.), Centre for Computational Medicine (M.B., N.S.), Division of Emergency Medicine (M.D.), and Division of Clinical and Metabolic Genetics (S.H.), Hospital for Sick Children; Princess Margaret Hospital (S.S.), Department of Medical Oncology and Hematology; University of Toronto (R.A.), Ontario, Canada; The Manton Center for Orphan Disease Research (A.H.B., C.A.G.), Division of Genetics and Genomics, Boston Children's Hospital, Harvard Medical School, MA; National Institute of Neurological Disorders and Stroke (C.G.B.), Neuromuscular and Neurogenetic Disorders of Childhood Section, and Clinical Research Center (M.S.J.), Rehabilitation Medicine Department, NIH, Bethesda, MD; Department of Computer Science (M.B., M.G., N.S.), University of Toronto, Ontario, Canada; Columbia University Irving Medical Center (A.C.), Division of Pediatric Pulmonology, New York, NY; Goryeb Children's Hospital (J.D.), Department of Pediatric Neurology, Morristown, NJ; Mount Sinai Hospital (C.H.), Prenatal Diagnosis and Medical Genetics, Toronto, Ontario, Canada; Medical College of Wisconsin (M.W.L.), Department of Pathology and Laboratory Medicine, Milwaukee; Children's Hospital of Philadelphia (O.H.M.), Division of Pulmonology, PA; UT Southwestern Medical Center (L.N.), Department of Physical Therapy, Dallas, TX; and Driscoll Children's Hospital (C.H.W.), Division of Neurology, Texas A&M University, Corpus Christi.
| |
Collapse
|
46
|
Sundaramurthy S, Votra S, Laszlo A, Davies T, Pruyne D. FHOD-1 is the only formin in Caenorhabditis elegans that promotes striated muscle growth and Z-line organization in a cell autonomous manner. Cytoskeleton (Hoboken) 2020; 77:422-441. [PMID: 33103378 DOI: 10.1002/cm.21639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 10/21/2020] [Accepted: 10/22/2020] [Indexed: 11/06/2022]
Abstract
The striated body wall muscles of Caenorhabditis elegans are a simple model for sarcomere assembly. Previously, we observed deletion mutants for two formin genes, fhod-1 and cyk-1, develop thin muscles with abnormal dense bodies (the sarcomere Z-line analogs). However, this work left in question whether these formins work in a muscle cell autonomous manner, particularly since cyk-1(∆) deletion has pleiotropic effects on development. Using a fast acting temperature-sensitive cyk-1(ts) mutant, we show here that neither postembryonic loss nor acute loss of CYK-1 during embryonic sarcomerogenesis cause lasting muscle defects. Furthermore, mosaic expression of CYK-1 in cyk-1(∆) mutants is unable to rescue muscle defects in a cell autonomous manner, suggesting muscle phenotypes caused by cyk-1(∆) are likely indirect. Conversely, mosaic expression of FHOD-1 in fhod-1(Δ) mutants promotes muscle cell growth and proper dense body organization in a muscle cell autonomous manner. As we observe no effect of loss of any other formin on muscle development, we conclude FHOD-1 is the only worm formin that directly promotes striated muscle development, and the effects on formin loss in C. elegans are surprisingly modest compared to other systems.
Collapse
Affiliation(s)
- Sumana Sundaramurthy
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - SarahBeth Votra
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Arianna Laszlo
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Tim Davies
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA.,Department of Biosciences, Durham University, Durham, UK
| | - David Pruyne
- Department of Cell and Developmental Biology, State University of New York Upstate Medical University, Syracuse, New York, USA
| |
Collapse
|
47
|
Lmod3 promotes myoblast differentiation and proliferation via the AKT and ERK pathways. Exp Cell Res 2020; 396:112297. [DOI: 10.1016/j.yexcr.2020.112297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/17/2020] [Accepted: 09/19/2020] [Indexed: 12/29/2022]
|
48
|
Tolkatchev D, Smith GE, Schultz LE, Colpan M, Helms GL, Cort JR, Gregorio CC, Kostyukova AS. Leiomodin creates a leaky cap at the pointed end of actin-thin filaments. PLoS Biol 2020; 18:e3000848. [PMID: 32898131 PMCID: PMC7500696 DOI: 10.1371/journal.pbio.3000848] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 09/18/2020] [Accepted: 08/17/2020] [Indexed: 01/26/2023] Open
Abstract
Improper lengths of actin-thin filaments are associated with altered contractile activity and lethal myopathies. Leiomodin, a member of the tropomodulin family of proteins, is critical in thin filament assembly and maintenance; however, its role is under dispute. Using nuclear magnetic resonance data and molecular dynamics simulations, we generated the first atomic structural model of the binding interface between the tropomyosin-binding site of cardiac leiomodin and the N-terminus of striated muscle tropomyosin. Our structural data indicate that the leiomodin/tropomyosin complex only forms at the pointed end of thin filaments, where the tropomyosin N-terminus is not blocked by an adjacent tropomyosin protomer. This discovery provides evidence supporting the debated mechanism where leiomodin and tropomodulin regulate thin filament lengths by competing for thin filament binding. Data from experiments performed in cardiomyocytes provide additional support for the competition model; specifically, expression of a leiomodin mutant that is unable to interact with tropomyosin fails to displace tropomodulin at thin filament pointed ends and fails to elongate thin filaments. Together with previous structural and biochemical data, we now propose a molecular mechanism of actin polymerization at the pointed end in the presence of bound leiomodin. In the proposed model, the N-terminal actin-binding site of leiomodin can act as a "swinging gate" allowing limited actin polymerization, thus making leiomodin a leaky pointed-end cap. Results presented in this work answer long-standing questions about the role of leiomodin in thin filament length regulation and maintenance.
Collapse
Affiliation(s)
- Dmitri Tolkatchev
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington, United States of America
| | - Garry E. Smith
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington, United States of America
| | - Lauren E. Schultz
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, United States of America
| | - Mert Colpan
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, United States of America
| | - Gregory L. Helms
- The Center for NMR Spectroscopy, Washington State University, Pullman, Washington, United States of America
| | - John R. Cort
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, Washington, United States of America
- Institute of Biological Chemistry, Washington State University, Pullman, Washington, United States of America
| | - Carol C. Gregorio
- Department of Cellular and Molecular Medicine and Sarver Molecular Cardiovascular Research Program, The University of Arizona, Tucson, Arizona, United States of America
| | - Alla S. Kostyukova
- Voiland School of Chemical Engineering and Bioengineering, Washington State University, Pullman, Washington, United States of America
| |
Collapse
|
49
|
Wang Q, Hu Z, Chang X, Yu M, Xie Z, Lv H, Zhang W, Xiong H, Yuan Y, Wang Z. Mutational and clinical spectrum in a cohort of Chinese patients with hereditary nemaline myopathy. Clin Genet 2020; 97:878-889. [PMID: 32222963 DOI: 10.1111/cge.13745] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 03/23/2020] [Accepted: 03/23/2020] [Indexed: 12/12/2022]
Abstract
Hereditary nemaline myopathy (NM) is one of the most common congenital myopathies with the histopathological findings of nemaline bodies. We used targeted next-generation sequencing to identify causative mutations in 48 NM patients with confirmed myopathological diagnosis, analyze the mutational spectrum and phenotypic features. Furthermore, reverse transcription polymerase chain reaction (RT-PCR) was used to confirm the pathogenic effect of one nebulin (NEB) splicing variant. The results showed that variants were found in five NM-associated genes, including NEB, actin alpha 1 (ACTA1), troponin T1, Kelch repeat and BTB domain-containing 13, and cofilin-2, in 34 (73.9%), 7 (15.2%), 3 (6.5%), 1 (2.2%), and 1 (2.2%) patients, respectively, in a total of 46/48 (95.8%) NM patients. Of the total 64 variants identified, 51 were novel variants including 26 pathogenic, 1 probably pathogenic, and 24 variant of uncertain significance (VUS). Notably, one NEB splicing mutation, c.21417+3A>G causing exon 144 splicing (NM_001164508.1), as confirmed by RT-PCR, was found in 52.9% (18 patients) of NEB variant-carrying patients. Typical congenital NM, the most common clinical subtype (60.4%), was associated with five NM genes. We concluded that hereditary NM showed a highly variable genetic spectrum. NEB was the most frequent causative gene in this Chinese cohort, followed by ACTA1. We found a hotspot splicing mutation in NEB among Chinese cohort.
Collapse
Affiliation(s)
- Qi Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhenxian Hu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Xingzhi Chang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhiying Xie
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - He Lv
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Hui Xiong
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
50
|
Laitila JM, McNamara EL, Wingate CD, Goullee H, Ross JA, Taylor RL, van der Pijl R, Griffiths LM, Harries R, Ravenscroft G, Clayton JS, Sewry C, Lawlor MW, Ottenheijm CAC, Bakker AJ, Ochala J, Laing NG, Wallgren-Pettersson C, Pelin K, Nowak KJ. Nebulin nemaline myopathy recapitulated in a compound heterozygous mouse model with both a missense and a nonsense mutation in Neb. Acta Neuropathol Commun 2020; 8:18. [PMID: 32066503 PMCID: PMC7027239 DOI: 10.1186/s40478-020-0893-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 02/05/2020] [Indexed: 12/31/2022] Open
Abstract
Nemaline myopathy (NM) caused by mutations in the gene encoding nebulin (NEB) accounts for at least 50% of all NM cases worldwide, representing a significant disease burden. Most NEB-NM patients have autosomal recessive disease due to a compound heterozygous genotype. Of the few murine models developed for NEB-NM, most are Neb knockout models rather than harbouring Neb mutations. Additionally, some models have a very severe phenotype that limits their application for evaluating disease progression and potential therapies. No existing murine models possess compound heterozygous Neb mutations that reflect the genotype and resulting phenotype present in most patients. We aimed to develop a murine model that more closely matched the underlying genetics of NEB-NM, which could assist elucidation of the pathogenetic mechanisms underlying the disease. Here, we have characterised a mouse strain with compound heterozygous Neb mutations; one missense (p.Tyr2303His), affecting a conserved actin-binding site and one nonsense mutation (p.Tyr935*), introducing a premature stop codon early in the protein. Our studies reveal that this compound heterozygous model, NebY2303H, Y935X, has striking skeletal muscle pathology including nemaline bodies. In vitro whole muscle and single myofibre physiology studies also demonstrate functional perturbations. However, no reduction in lifespan was noted. Therefore, NebY2303H,Y935X mice recapitulate human NEB-NM and are a much needed addition to the NEB-NM mouse model collection. The moderate phenotype also makes this an appropriate model for studying NEB-NM pathogenesis, and could potentially be suitable for testing therapeutic applications.
Collapse
|