1
|
Shimazaki R, Saito Y, Awaya T, Minami N, Kurosawa R, Hosokawa M, Ohara H, Hayashi S, Takeuchi A, Hagiwara M, Hayashi YK, Noguchi S, Nishino I. Profiling of pathogenic variants in Japanese patients with sarcoglycanopathy. Orphanet J Rare Dis 2025; 20:1. [PMID: 39755676 DOI: 10.1186/s13023-024-03521-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 12/26/2024] [Indexed: 01/06/2025] Open
Abstract
BACKGROUND Sarcoglycanopathies (SGPs) are limb-girdle muscular dystrophies (LGMDs) that can be classified into four types, LGMDR3, LGMDR4, LGMDR5, and LGMDR6, caused by mutations in the genes, SGCA, SGCB, SGCG, and SGCD, respectively. SGPs are relatively rare in Japan. This study aims to profile the genetic variants that cause SGPs in Japanese patients. METHODS Clinical course and pathological findings were retrospectively reviewed in Japanese patients with SGP. Genetic analyses were performed using a combination of targeted resequencing with a hereditary muscle disease panel, whole genome sequencing, multiplex ligation-dependent probe amplification, and long-read sequencing. The structures of transcripts with aberrant splicing were also determined by RT-PCR, RNA-seq, and in silico prediction. RESULTS We identified biallelic variants in SGC genes in 53 families, including three families with LGMDR6, which had not been identified in Japan so far. SGCA was the most common causative gene, accounting for 56% of cases, followed by SGCG, SGCB, and SGCD, at 17%, 21%, and 6%, respectively. Missense variants in SGCA were very frequent at 78.3%, while they were relatively rare in SGCB, SGCG, and SGCD at 11.1%, 18.2%, and 16.6%, respectively. We also analyzed the haplotypes of alleles carrying three variants found in multiple cases: c.229C > T in SGCA, c.325C > T in SGCB, and exon 6 deletion in SGCG; two distinct haplotypes were found for c.229C > T in SGCA, while each of the latter two variants was on single haplotypes. CONCLUSIONS We present genetic profiles of Japanese patients with SGPs. Haplotype analysis indicated common ancestors of frequent variants. Our findings will support genetic diagnosis and gene therapy.
Collapse
Affiliation(s)
- Rui Shimazaki
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Yoshihiko Saito
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan
- Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Tomonari Awaya
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
- Center for Anatomical Studies, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Narihiro Minami
- Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ryo Kurosawa
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Motoyasu Hosokawa
- Department of Developmental Biology and Functional Genomics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Hiroaki Ohara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Shinichiro Hayashi
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan
| | - Akihide Takeuchi
- Department of Developmental Biology and Functional Genomics, Ehime University Graduate School of Medicine, Ehime, Japan
| | - Masatoshi Hagiwara
- Department of Anatomy and Developmental Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yukiko K Hayashi
- Department of Pathophysiology, Tokyo Medical University, Tokyo, Japan
| | - Satoru Noguchi
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan.
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, 4-1-1 Ogawa-Higashi, Kodaira, Tokyo, 187-8502, Japan
- Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| |
Collapse
|
2
|
Wan L, Ge X, Xu Q, Huang G, Yang T, Campbell KP, Yan Z, Wu J. Structure and assembly of the dystrophin glycoprotein complex. Nature 2025; 637:1252-1260. [PMID: 39663450 DOI: 10.1038/s41586-024-08310-2] [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: 06/13/2024] [Accepted: 10/30/2024] [Indexed: 12/13/2024]
Abstract
The dystrophin glycoprotein complex (DGC) has a crucial role in maintaining cell membrane stability and integrity by connecting the intracellular cytoskeleton with the surrounding extracellular matrix1-3. Dysfunction of dystrophin and its associated proteins results in muscular dystrophy, a disorder characterized by progressive muscle weakness and degeneration4,5. Despite the important roles of the DGC in physiology and pathology, its structural details remain largely unknown, hindering a comprehensive understanding of its assembly and function. Here we isolated the native DGC from mouse skeletal muscle and obtained its high-resolution structure. Our findings unveil a markedly divergent structure from the previous model of DGC assembly. Specifically, on the extracellular side, β-, γ- and δ-sarcoglycans co-fold to form a specialized, extracellular tower-like structure, which has a central role in complex assembly by providing binding sites for α-sarcoglycan and dystroglycan. In the transmembrane region, sarcoglycans and sarcospan flank and stabilize the single transmembrane helix of dystroglycan, rather than forming a subcomplex as previously proposed6-8. On the intracellular side, sarcoglycans and dystroglycan engage in assembly with the dystrophin-dystrobrevin subcomplex through extensive interaction with the ZZ domain of dystrophin. Collectively, these findings enhance our understanding of the structural linkage across the cell membrane and provide a foundation for the molecular interpretation of many muscular dystrophy-related mutations.
Collapse
Affiliation(s)
- Li Wan
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Xiaofei Ge
- State Key Laboratory of Membrane Biology, Beijing Frontier Research Center for Biological Structure, School of Life Sciences, Tsinghua University, Beijing, China
| | - Qikui Xu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Gaoxingyu Huang
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China
| | - Tiandi Yang
- Howard Hughes Medical Institute, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
- Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
| | - Kevin P Campbell
- Howard Hughes Medical Institute, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
- Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
- Department of Molecular Physiology and Biophysics, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
- Department of Neurology, University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA, USA
| | - Zhen Yan
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China.
| | - Jianping Wu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, China.
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, China.
- Institute of Biology, Westlake Institute for Advanced Study, Hangzhou, China.
| |
Collapse
|
3
|
Rahmuni Y, Kadiri YE, Lyahyai J, Sefiani A, Ratbi I. Molecular diagnosis of Alpha-sarcoglycanopathies by NGS in seven Moroccan families and report of two novel variants. Ir J Med Sci 2024; 193:3071-3076. [PMID: 39174842 DOI: 10.1007/s11845-024-03792-5] [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: 04/01/2024] [Accepted: 08/18/2024] [Indexed: 08/24/2024]
Abstract
BACKGROUND Limb-girdle muscular dystrophies constitute a heterogeneous group of neuromuscular diseases, both clinically and genetically. Limb-girdle muscular dystrophy by alpha-sarcoglycan deficiency or LGMD R3 α-sarcoglycan-related is a subtype of the autosomal recessive sarcoglycanopathies caused by variants in the alpha-sarcoglycan gene (SGCA) at 17q21.33. It appears in childhood by progressive weakness of pelvic and/or scapular girdle muscles and calf hypertrophy, with a wide range of clinical inter- and intra-familial clinical variability. AIMS Our report extends the molecular spectrum of SGCA gene with the identification of variant disease causing and will help for better management of patients and genetic counseling of families. METHODS In our study, seven unrelated families presented a clinical and paraclinical picture consistent with alpha-sarcoglycanopathy. A molecular study using Next-Generation Sequencing (NGS) was carried out on them. RESULTS Six different homozygous variants of the SGCA gene were identified in the patients analyzed, including four previously reported variants and two novel variants predicted to be deleterious by the prediction tools. CONCLUSIONS Our results expand the spectrum of variants in Moroccan patients with sarcoglycanopathy, specifically LGMDR3, most importantly as this form is not common in the Moroccan population.
Collapse
Affiliation(s)
- Yasmina Rahmuni
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, GENOPATH Center, Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco.
- Department of Medical Genetics, National Institute of Health, Rabat, Morocco.
| | - Youssef El Kadiri
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, GENOPATH Center, Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco
- Department of Medical Genetics, National Institute of Health, Rabat, Morocco
| | - Jaber Lyahyai
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, GENOPATH Center, Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco
| | - Abdelaziz Sefiani
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, GENOPATH Center, Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco
- Department of Medical Genetics, National Institute of Health, Rabat, Morocco
| | - Ilham Ratbi
- Research Team in Genomics and Molecular Epidemiology of Genetic Diseases, GENOPATH Center, Faculty of Medicine and Pharmacy, University Mohammed V of Rabat, Rabat, Morocco
| |
Collapse
|
4
|
Bulakh M, Polyakova D, Dadali E, Rudenskaya G, Sharkova I, Markova T, Murtazina A, Demina N, Kurbatov S, Nikitina N, Udalova V, Polyakov A, Ryzhkova O. Genetic spectrum of sarcoglycanopathies in a cohort of Russian patients. Gene 2024; 927:148680. [PMID: 38876406 DOI: 10.1016/j.gene.2024.148680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 06/06/2024] [Accepted: 06/10/2024] [Indexed: 06/16/2024]
Abstract
Sarcoglycanopathies encompass four distinct forms of limb-girdle muscular dystrophies (LGMD), denoted as LGMD R3-R6, arising from mutations within the SGCA, SGCB, SGCG, and SGCD genes. The global prevalence of sarcoglycanopathies is low, making it challenging to study these diseases. The principal objective of this study was to explore the spectrum of mutations in a cohort of Russian patients with sarcoglycanopathies and to ascertain the frequency of these conditions in the Russian Federation. We conducted a retrospective analysis of clinical and molecular genetic data from 49 Russian patients with sarcoglycan genes variants. The results indicated that variants in the SGCA gene were found in 71.4% of cases, with SGCB and SGCG genes each exhibiting variants in 12.2 % of patients. SGCD gene variants were detected in 4.1% of cases. Bi-allelic pathogenic and likely pathogenic variants were identified in 46 of the 49 cases of sarcoglycanopathies: LGMD R3 (n = 34), LGMD R4 (n = 4), LGMD R5 (n = 6), and LGMD R6 (n = 2). A total of 31 distinct variants were identified, comprising 25 previously reported and 6 novel variants. Two major variants, c.229C>T and c.271G>A, were detected within the SGCA, constituting 61.4% of all mutant alleles in Russian patients with LGMD R3. Both LGMD R6 cases were caused by the homozygous nonsense variant c.493C>T p.(Arg165Ter) in the SGCD gene. The incidence of sarcoglycanopathies in the Russian Federation was estimated to be at least 1 in 4,115,039, which is lower than the reported incidence in other populations.
Collapse
Affiliation(s)
- Maria Bulakh
- Research Centre for Medical Genetics, Moscow, Russia.
| | | | - Elena Dadali
- Research Centre for Medical Genetics, Moscow, Russia.
| | | | - Inna Sharkova
- Research Centre for Medical Genetics, Moscow, Russia.
| | | | | | - Nina Demina
- Research Centre for Medical Genetics, Moscow, Russia.
| | - Sergei Kurbatov
- Research Institute of Experimental Biology and Medicine, Voronezh State Medical University N.N. Burdenko, Voronezh, Russia; Saratov State Medical University, Saratov, Russia.
| | - Natalia Nikitina
- State Healthcare Institution of Sverdlovsk Region "Clinical Diagnostic Center "Mother's and Child Health Protection", Yekaterinburg, Russia.
| | | | | | | |
Collapse
|
5
|
Clinical, genetic profile and disease progression of sarcoglycanopathies in a large cohort from India: high prevalence of SGCB c.544A > C. Neurogenetics 2022; 23:187-202. [DOI: 10.1007/s10048-022-00690-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/08/2022] [Indexed: 10/18/2022]
|
6
|
Hoch L, Bourg N, Degrugillier F, Bruge C, Benabides M, Pellier E, Tournois J, Mahé G, Maignan N, Dawe J, Georges M, Papazian D, Subramanian N, Simon S, Fanen P, Delevoye C, Richard I, Nissan X. Dual Blockade of Misfolded Alpha-Sarcoglycan Degradation by Bortezomib and Givinostat Combination. Front Pharmacol 2022; 13:856804. [PMID: 35571097 PMCID: PMC9093689 DOI: 10.3389/fphar.2022.856804] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
Limb-girdle muscular dystrophy type R3 (LGMD R3) is a rare genetic disorder characterized by a progressive proximal muscle weakness and caused by mutations in the SGCA gene encoding alpha-sarcoglycan (α-SG). Here, we report the results of a mechanistic screening ascertaining the molecular mechanisms involved in the degradation of the most prevalent misfolded R77C-α-SG protein. We performed a combinatorial study to identify drugs potentializing the effect of a low dose of the proteasome inhibitor bortezomib on the R77C-α-SG degradation inhibition. Analysis of the screening associated to artificial intelligence-based predictive ADMET characterization of the hits led to identification of the HDAC inhibitor givinostat as potential therapeutical candidate. Functional characterization revealed that givinostat effect was related to autophagic pathway inhibition, unveiling new theories concerning degradation pathways of misfolded SG proteins. Beyond the identification of a new therapeutic option for LGMD R3 patients, our results shed light on the potential repurposing of givinostat for the treatment of other genetic diseases sharing similar protein degradation defects such as LGMD R5 and cystic fibrosis.
Collapse
Affiliation(s)
- Lucile Hoch
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | - Nathalie Bourg
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, Evry, France
| | | | - Céline Bruge
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | - Manon Benabides
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | - Emilie Pellier
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | - Johana Tournois
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | - Gurvan Mahé
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| | | | | | | | | | | | | | - Pascale Fanen
- Université Paris Est Creteil, INSERM, IMRB, Créteil, France.,Département de Genetique, DMU Biologie-Pathologie, GH Mondor-A. Chenevier, AP-HP, Creteil, France
| | - Cédric Delevoye
- Institut Curie, PSL Research University, CNRS, UMR144, Structure and Membrane Compartments, Paris, France.,Institut Curie, PSL Research University, CNRS, UMR144, Cell and Tissue Imaging Facility (PICT-IBiSA), Paris, France
| | - Isabelle Richard
- INTEGRARE, Genethon, Inserm, Univ Evry, Université Paris-Saclay, Evry, France
| | - Xavier Nissan
- CECS, I-Stem, Corbeil-Essonne, France.,INSERM U861, I-Stem, Corbeil-Essonne, France.,UEVE U861, I-Stem, Corbeil-Essonne, France
| |
Collapse
|
7
|
Carson L, Merrick D. Genotype-phenotype correlations in alpha-sarcoglycanopathy: a systematic review. Ir J Med Sci 2022; 191:2743-2750. [PMID: 35040091 DOI: 10.1007/s11845-021-02855-1] [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: 03/18/2021] [Accepted: 10/27/2021] [Indexed: 11/27/2022]
Abstract
BACKGROUND Mutations in the alpha-sarcoglycan gene cause limb-girdle muscular dystrophy 2D, an autosomal recessive muscle wasting disorder primarily affecting the muscles of the shoulder and pelvic girdles. To date, no previous study has collated all known mutations in alpha-sarcoglycan and mapped these to the associated phenotypes. AIMS To examine for correlations between mutation locations, or mutation type, and the phenotype caused in all reported mutations in alpha-sarcoglycan. METHODS We present a systematic literature review examining correlations between mutation locations, or mutation type, and the phenotype caused in all reported cases of limb-girdle muscular dystrophy 2D. RESULTS From 134 unique genotypes collated, a strong prevalence of missense mutations (64% of all unique mutations) was found in this gene. Mutation hotspots were noted in exon three and the extracellular domain, with mutation densities varying significantly between both exons and protein domains (p ≤ 0.01). All compound heterozygous limb-girdle muscular dystrophy 2D patients with cardiac involvement contained at least one mutation in exon three, a novel finding. All non-sense mutations in alpha-sarcoglycan give a severe phenotype, as do genotypes involving a combination of exons four and five. This study confirms on a large, diverse cohort the extremely high prevalence of the c.229C > T mutation. CONCLUSIONS This study demonstrates the vast variation in disease severity seen between patients possessing the same mutation, highlighting the difficulty identifying genotype-phenotype correlations in this condition. Novel findings including the involvement of exon three in all compound heterozygous patients who suffered from cardiomyopathy, and the severity of mutations involving exons four and five may help to guide investigations and therapeutic decisions in an era of personalised medicine.
Collapse
Affiliation(s)
- Luke Carson
- School of Life Sciences, University of Nottingham, Nottingham, UK.
| | - Deborah Merrick
- School of Life Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
8
|
Abstract
The limb-girdle muscular dystrophies (LGMD) are a collection of genetic diseases united in their phenotypical expression of pelvic and shoulder area weakness and wasting. More than 30 subtypes have been identified, five dominant and 26 recessive. The increase in the characterization of new genotypes in the family of LGMDs further adds to the heterogeneity of the disease. Meanwhile, better understanding of the phenotype led to the reconsideration of the disease definition, which resulted in eight old subtypes to be no longer recognized officially as LGMD and five new diseases to be added to the LGMD family. The unique variabilities of LGMD stem from genetic mutations, which then lead to protein and ultimately muscle dysfunction. Herein, we review the LGMD pathway, starting with the genetic mutations that encode proteins involved in muscle maintenance and repair, and including the genotype–phenotype relationship of the disease, the epidemiology, disease progression, burden of illness, and emerging treatments.
Collapse
|
9
|
Escobar H, Krause A, Keiper S, Kieshauer J, Müthel S, de Paredes MG, Metzler E, Kühn R, Heyd F, Spuler S. Base editing repairs an SGCA mutation in human primary muscle stem cells. JCI Insight 2021; 6:145994. [PMID: 33848270 PMCID: PMC8262330 DOI: 10.1172/jci.insight.145994] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 04/07/2021] [Indexed: 11/28/2022] Open
Abstract
Skeletal muscle can regenerate from muscle stem cells and their myogenic precursor cell progeny, myoblasts. However, precise gene editing in human muscle stem cells for autologous cell replacement therapies of untreatable genetic muscle diseases has not yet been reported. Loss-of-function mutations in SGCA, encoding α-sarcoglycan, cause limb-girdle muscular dystrophy 2D/R3, an early-onset, severe, and rapidly progressive form of muscular dystrophy affecting both male and female patients. Patients suffer from muscle degeneration and atrophy affecting the limbs, respiratory muscles, and heart. We isolated human muscle stem cells from 2 donors, with the common SGCA c.157G>A mutation affecting the last coding nucleotide of exon 2. We found that c.157G>A is an exonic splicing mutation that induces skipping of 2 coregulated exons. Using adenine base editing, we corrected the mutation in the cells from both donors with > 90% efficiency, thereby rescuing the splicing defect and α-sarcoglycan expression. Base-edited patient cells regenerated muscle and contributed to the Pax7+ satellite cell compartment in vivo in mouse xenografts. Here, we provide the first evidence to our knowledge that autologous gene–repaired human muscle stem cells can be harnessed for cell replacement therapies of muscular dystrophies.
Collapse
Affiliation(s)
- Helena Escobar
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany.,Charité Universitätsmedizin Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Anne Krause
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany.,Charité Universitätsmedizin Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Sandra Keiper
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of RNA Biochemistry, Berlin, Germany
| | - Janine Kieshauer
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany.,Charité Universitätsmedizin Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Stefanie Müthel
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany.,Charité Universitätsmedizin Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Manuel García de Paredes
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany
| | - Eric Metzler
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany.,Charité Universitätsmedizin Berlin, Germany.,Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Ralf Kühn
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Florian Heyd
- Freie Universität Berlin, Institute of Chemistry and Biochemistry, Laboratory of RNA Biochemistry, Berlin, Germany
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, a cooperation between the Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association and the Charité, Universitätsmedizin Berlin, Germany
| |
Collapse
|
10
|
Chakravorty S, Nallamilli BRR, Khadilkar SV, Singla MB, Bhutada A, Dastur R, Gaitonde PS, Rufibach LE, Gloster L, Hegde M. Clinical and Genomic Evaluation of 207 Genetic Myopathies in the Indian Subcontinent. Front Neurol 2020; 11:559327. [PMID: 33250842 PMCID: PMC7674836 DOI: 10.3389/fneur.2020.559327] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/23/2020] [Indexed: 12/13/2022] Open
Abstract
Objective: Inherited myopathies comprise more than 200 different individually rare disease-subtypes, but when combined together they have a high prevalence of 1 in 6,000 individuals across the world. Our goal was to determine for the first time the clinical- and gene-variant spectrum of genetic myopathies in a substantial cohort study of the Indian subcontinent. Methods: In this cohort study, we performed the first large clinical exome sequencing (ES) study with phenotype correlation on 207 clinically well-characterized inherited myopathy-suspected patients from the Indian subcontinent with diverse ethnicities. Results: Clinical-correlation driven definitive molecular diagnosis was established in 49% (101 cases; 95% CI, 42–56%) of patients with the major contributing pathogenicity in either of three genes, GNE (28%; GNE-myopathy), DYSF (25%; Dysferlinopathy), and CAPN3 (19%; Calpainopathy). We identified 65 variant alleles comprising 37 unique variants in these three major genes. Seventy-eight percent of the DYSF patients were homozygous for the detected pathogenic variant, suggesting the need for carrier-testing for autosomal-recessive disorders like Dysferlinopathy that are common in India. We describe the observed clinical spectrum of myopathies including uncommon and rare subtypes in India: Sarcoglycanopathies (SGCA/B/D/G), Collagenopathy (COL6A1/2/3), Anoctaminopathy (ANO5), telethoninopathy (TCAP), Pompe-disease (GAA), Myoadenylate-deaminase-deficiency-myopathy (AMPD1), myotilinopathy (MYOT), laminopathy (LMNA), HSP40-proteinopathy (DNAJB6), Emery-Dreifuss-muscular-dystrophy (EMD), Filaminopathy (FLNC), TRIM32-proteinopathy (TRIM32), POMT1-proteinopathy (POMT1), and Merosin-deficiency-congenital-muscular-dystrophy-type-1 (LAMA2). Thirteen patients harbored pathogenic variants in >1 gene and had unusual clinical features suggesting a possible role of synergistic-heterozygosity/digenic-contribution to disease presentation and progression. Conclusions: Application of clinically correlated ES to myopathy diagnosis has improved our understanding of the clinical and genetic spectrum of different subtypes and their overlaps in Indian patients. This, in turn, will enhance the global gene-variant-disease databases by including data from developing countries/continents for more efficient clinically driven molecular diagnostics.
Collapse
Affiliation(s)
- Samya Chakravorty
- Emory University Department of Pediatrics, Atlanta, GA, United States.,Emory University Department of Human Genetics, Atlanta, GA, United States.,Division of Neurosciences, Children's Healthcare of Atlanta, Atlanta, GA, United States.,School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | | | - Satish Vasant Khadilkar
- Department of Neurology, Bombay Hospital, Mumbai, India.,Department of Neurology, Sir J J Group of Hospitals, Grant Medical College, Mumbai, India.,Bombay Hospital Institute of Medical Sciences, Mumbai, India
| | - Madhu Bala Singla
- Department of Neurology, Bombay Hospital, Mumbai, India.,Department of Neurology, Sir J J Group of Hospitals, Grant Medical College, Mumbai, India.,Bombay Hospital Institute of Medical Sciences, Mumbai, India
| | | | - Rashna Dastur
- Centre for Advanced Molecular Diagnostics in Neuromuscular Disorders (CAMDND), Mumbai, India
| | - Pradnya Satish Gaitonde
- Centre for Advanced Molecular Diagnostics in Neuromuscular Disorders (CAMDND), Mumbai, India
| | | | - Logan Gloster
- Emory University Department of Pediatrics, Atlanta, GA, United States.,School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States
| | - Madhuri Hegde
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, United States.,PerkinElmer Genomics, Global Laboratory Services, Waltham, MA, United States
| |
Collapse
|
11
|
Beecroft SJ, Yau KS, Allcock RJN, Mina K, Gooding R, Faiz F, Atkinson VJ, Wise C, Sivadorai P, Trajanoski D, Kresoje N, Ong R, Duff RM, Cabrera‐Serrano M, Nowak KJ, Pachter N, Ravenscroft G, Lamont PJ, Davis MR, Laing NG. Targeted gene panel use in 2249 neuromuscular patients: the Australasian referral center experience. Ann Clin Transl Neurol 2020; 7:353-362. [PMID: 32153140 PMCID: PMC7086001 DOI: 10.1002/acn3.51002] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 01/28/2020] [Accepted: 02/06/2020] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE To develop, test, and iterate a comprehensive neuromuscular targeted gene panel in a national referral center. METHODS We designed two iterations of a comprehensive targeted gene panel for neuromuscular disorders. Version 1 included 336 genes, which was increased to 464 genes in Version 2. Both panels used TargetSeqTM probe-based hybridization for target enrichment followed by Ion Torrent sequencing. Targeted high-coverage sequencing and analysis was performed on 2249 neurology patients from Australia and New Zealand (1054 Version 1, 1195 Version 2) from 2012 to 2015. No selection criteria were used other than referral from a suitable medical specialist (e.g., neurologist or clinical geneticist). Patients were classified into 15 clinical categories based on the clinical diagnosis from the referring clinician. RESULTS Six hundred and sixty-five patients received a genetic diagnosis (30%). Diagnosed patients were significantly younger that undiagnosed patients (26.4 and 32.5 years, respectively; P = 4.6326E-9). The diagnostic success varied markedly between disease categories. Pathogenic variants in 10 genes explained 38% of the disease burden. Unexpected phenotypic expansions were discovered in multiple cases. Triage of unsolved cases for research exome testing led to the discovery of six new disease genes. INTERPRETATION A comprehensive targeted diagnostic panel was an effective method for neuromuscular disease diagnosis within the context of an Australasian referral center. Use of smaller disease-specific panels would have precluded diagnosis in many patients and increased cost. Analysis through a centralized laboratory facilitated detection of recurrent, but under-recognized pathogenic variants.
Collapse
Affiliation(s)
- Sarah J. Beecroft
- Centre for Medical ResearchUniversity of Western AustraliaHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Kyle S. Yau
- Centre for Medical ResearchUniversity of Western AustraliaHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Richard J. N. Allcock
- School of Biomedical SciencesUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Kym Mina
- Department of Diagnostic GenomicsDepartment of HealthPathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Rebecca Gooding
- Department of Diagnostic GenomicsDepartment of HealthPathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Fathimath Faiz
- Department of Diagnostic GenomicsDepartment of HealthPathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Vanessa J. Atkinson
- School of Biomedical SciencesUniversity of Western AustraliaPerthWestern AustraliaAustralia
- Department of Diagnostic GenomicsDepartment of HealthPathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Cheryl Wise
- Department of Diagnostic GenomicsDepartment of HealthPathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Padma Sivadorai
- Department of Diagnostic GenomicsDepartment of HealthPathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Daniel Trajanoski
- Department of Diagnostic GenomicsDepartment of HealthPathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Nina Kresoje
- School of Biomedical SciencesUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Royston Ong
- Centre for Medical ResearchUniversity of Western AustraliaHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Rachael M. Duff
- Centre for Medical ResearchUniversity of Western AustraliaHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Macarena Cabrera‐Serrano
- Department of NeurologyHospital Universitario Virgen del RocioInstituto de Biomedicina de SevillaCSICUniversidad de SevillaSevillaSpain
| | - Kristen J. Nowak
- Centre for Medical ResearchUniversity of Western AustraliaHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsWestern AustraliaAustralia
- Public and Aboriginal Health DivisionDepartment of HealthOffice of Population Health GenomicsPerthWestern AustraliaAustralia
| | - Nicholas Pachter
- Genetic Services of Western AustraliaDepartment of HealthGovernment of Western AustraliaPerthWestern AustraliaAustralia
- School of Paediatrics and Child HealthUniversity of Western AustraliaPerthWestern AustraliaAustralia
| | - Gianina Ravenscroft
- Centre for Medical ResearchUniversity of Western AustraliaHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsWestern AustraliaAustralia
| | | | - Mark R. Davis
- Department of Diagnostic GenomicsDepartment of HealthPathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| | - Nigel G. Laing
- Centre for Medical ResearchUniversity of Western AustraliaHarry Perkins Institute of Medical ResearchQEII Medical CentreNedlandsWestern AustraliaAustralia
- Department of Diagnostic GenomicsDepartment of HealthPathWest Laboratory MedicineQEII Medical CentreNedlandsWestern AustraliaAustralia
| |
Collapse
|
12
|
Oliveira Santos M, Coelho P, Roque R, Conceição I. Very late-onset limb-girdle muscular dystrophy type 2D: A milder form with a normal muscle biopsy. J Clin Neurosci 2019; 72:471-473. [PMID: 31836381 DOI: 10.1016/j.jocn.2019.12.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 12/01/2019] [Indexed: 11/18/2022]
Abstract
Sarcoglycanopathies are a genetically heterogeneous group of autosomal recessive limb-girdle muscular dystrophies (LGMD) caused by mutations in sarcoglycan genes. We report a Portuguese patient with a very late-onset LGMD phenotype, whose muscle biopsy and immunostaining, in particular for α-sarcoglycan, were unrevealing. Muscle MRI showed a predominant, bilateral and symmetric involvement of the tight muscles and also, to a lesser extent, of the posterior compartment of lower legs muscles. Next generation sequencing (NGS) revealed a known homozygous c.850C > T (p.Arg284Cys) mutation in SGCA gene. Milder forms of α-sarcoglycanopathies could be a challenging diagnosis; particularly if muscle histopathology and α-sarcoglycan immunohistochemistry are unhelpful. NGS plays a crucial role not only for aiding in the establishment of a definite diagnosis, but also for expanding clinical presentations.
Collapse
Affiliation(s)
- Miguel Oliveira Santos
- Department of Neurosciences and Mental Health, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal; Institute of Physiology, Instituto de Medicina Molecular, Faculty of Medicine, University of Lisbon, Lisbon, Portugal.
| | - Pedro Coelho
- Department of Neurosciences and Mental Health, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal
| | - Rafael Roque
- Department of Neurosciences and Mental Health, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal
| | - Isabel Conceição
- Department of Neurosciences and Mental Health, Hospital de Santa Maria, Centro Hospitalar Universitário de Lisboa Norte, Lisbon, Portugal; Institute of Physiology, Instituto de Medicina Molecular, Faculty of Medicine, University of Lisbon, Lisbon, Portugal
| |
Collapse
|
13
|
Özyilmaz B, Kirbiyik Ö, Özdemir TR, Kaya Özer Ö, Kutbay YB, Erdogan KM, Güvenç MS, Kale MY, Gazeteci H, Kiliç B, Sertpoyraz F, Diniz G, Baydan F, Gençpinar P, Dündar NO, Yiş U. Impact of next‐generation sequencing panels in the evaluation of limb‐girdle muscular dystrophies. Ann Hum Genet 2019; 83:331-347. [DOI: 10.1111/ahg.12319] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2019] [Revised: 04/04/2019] [Accepted: 04/09/2019] [Indexed: 12/23/2022]
Affiliation(s)
- Berk Özyilmaz
- Genetic Diagnosis Center, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
- Neuromuscular Disorders Unit, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
| | - Özgür Kirbiyik
- Genetic Diagnosis Center, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
| | - Taha R. Özdemir
- Genetic Diagnosis Center, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
| | - Özge Kaya Özer
- Genetic Diagnosis Center, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
| | - Yaşar B. Kutbay
- Genetic Diagnosis Center, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
| | - Kadri M. Erdogan
- Genetic Diagnosis Center, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
| | - Merve Saka Güvenç
- Genetic Diagnosis Center, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
| | - Murat Yildirim Kale
- Neuromuscular Disorders Unit, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
| | - Hande Gazeteci
- Pediatric Neurology Cigli District Training Hospital Izmir Turkey
| | - Betül Kiliç
- Pediatric Neurology Derince Education Research Hospital Kocaeli Turkey
| | - Filiz Sertpoyraz
- Neuromuscular Disorders Unit, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
| | - Gulden Diniz
- Department of Pathology Izmir Democracy University Izmir Turkey
| | - Figen Baydan
- Neuromuscular Disorders Unit, Tepecik Training and Research Hospital University of Health Sciences Izmir Turkey
| | - Pinar Gençpinar
- Pediatric Neurology, Faculty of Medicine Izmir Katip Celebi University Izmir Turkey
| | - Nihal Olgaç Dündar
- Pediatric Neurology, Faculty of Medicine Izmir Katip Celebi University Izmir Turkey
| | - Uluç Yiş
- Pediatric Neurology, School of Medicine Dokuz Eylül University Izmir Turkey
| |
Collapse
|
14
|
Identification of thiostrepton as a pharmacological approach to rescue misfolded alpha-sarcoglycan mutant proteins from degradation. Sci Rep 2019; 9:6915. [PMID: 31061434 PMCID: PMC6502821 DOI: 10.1038/s41598-019-43399-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2018] [Accepted: 04/18/2019] [Indexed: 12/12/2022] Open
Abstract
Limb-girdle muscular dystrophy type 2D (LGMD2D) is characterized by a progressive proximal muscle weakness. LGMD2D is caused by mutations in the gene encoding α-sarcoglycan (α-SG), a dystrophin-associated glycoprotein that plays a key role in the maintenance of sarcolemma integrity in striated muscles. We report here on the development of a new in vitro high-throughput screening assay that allows the monitoring of the proper localization of the most prevalent mutant form of α-SG (R77C substitution). Using this assay, we screened a library of 2560 FDA-approved drugs and bioactive compounds and identified thiostrepton, a cyclic antibiotic, as a potential drug to repurpose for LGMD2D treatment. Characterization of the thiostrepton effect revealed a positive impact on R77C-α-SG and other missense mutant protein localization (R34H, I124T, V247M) in fibroblasts overexpressing these proteins. Finally, further investigations of the molecular mechanisms of action of the compound revealed an inhibition of the chymotrypsin-like activity of the proteasome 24 h after thiostrepton treatment and a synergistic effect with bortezomib, an FDA-approved proteasome inhibitor. This study reports on the first in vitro model for LGMD2D that is compatible with high-throughput screening and proposes a new therapeutic option for LGMD2D caused by missense mutations of α-SG.
Collapse
|
15
|
Lu Y, Song X, Ji G, Wu H, Li D, Sun S. Identification of a novel SGCA missense mutation in a case of limb-girdle muscular dystrophy 2D with the absence of four sarcoglycan proteins. Neuropathology 2019; 39:207-211. [PMID: 30989758 PMCID: PMC6850699 DOI: 10.1111/neup.12549] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 02/18/2019] [Accepted: 02/18/2019] [Indexed: 12/02/2022]
Abstract
Limb‐girdle muscular dystrophy 2D (LGMD2D) is caused by mutations in the α‐sarcoglycan gene (SGCA). Due to lack of specificity, it is impossible to identify LGMD2D only by clinical symptoms and conventional immunohistochemical staining. The loss of any protein (α‐, β‐, γ‐, δ‐sarcoglycan) that represent sarcoglycanopathy may cause reduction or absence of the other three proteins. Here, we report a patient with a complete loss of all the four proteins. Next generation sequencing (NGS) results showed a missense mutation (C.218 C > T) and a partial heterozygous deletion containing exons 7 and 8 of SGCA, which led to the final diagnosis of the patient. The discovery of this new mutation could broaden the spectrum of SGCA mutations, which may be associated with putative LGMD2D, especially when all the four proteins are completely missing.
Collapse
Affiliation(s)
- Yanpeng Lu
- Department of Neurology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Xueqin Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Guang Ji
- Department of Neurology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Hongran Wu
- Department of Neurology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Duan Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Hebei, China
| | - Shuyan Sun
- Department of Neurology, The Second Hospital of Hebei Medical University, Hebei, China
| |
Collapse
|
16
|
Xie Z, Hou Y, Yu M, Liu Y, Fan Y, Zhang W, Wang Z, Xiong H, Yuan Y. Clinical and genetic spectrum of sarcoglycanopathies in a large cohort of Chinese patients. Orphanet J Rare Dis 2019; 14:43. [PMID: 30764848 PMCID: PMC6376703 DOI: 10.1186/s13023-019-1021-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2018] [Accepted: 02/03/2019] [Indexed: 11/20/2022] Open
Abstract
Background Sarcoglycanopathies comprise four subtypes of autosomal recessive limb-girdle muscular dystrophy (LGMD2C, LGMD2D, LGMD2E, and LGMD2F) that are caused, respectively, by mutations in the SGCG, SGCA, SGCB, and SGCD genes. Knowledge about the clinical and genetic features of sarcoglycanopathies in Chinese patients is limited. The aims of this study were to investigate in detail the clinical manifestations, sarcoglycan expression, and gene mutations in Chinese patients with sarcoglycanopathies and to identify possible correlations between them. Results Of 3638 patients for suspected neuromuscular diseases (1733 with inherited myopathies, 1557 with acquired myopathies, and 348 unknown), 756 patients had next-generation sequencing (NGS) diagnostic panel. Twenty-five patients with sarcoglycanopathies (11.5%) were identified from 218 confirmed LGMDs, comprising 18 with LGMD2D, 6 with LGMD2E, and one with LGMD2C. One patient with LGMD2D also had Charcot-Marie-Tooth 1A. The clinical phenotypes of the patients with LGMD2D or LGMD2E were markedly heterogeneous. Muscle biopsy showed a dystrophic pattern in 19 patients and mild myopathic changes in 6. The percentage of correct prediction of genotype based on expression of sarcoglycan was 36.0% (4 LGMD2D, 4 LGMD2E, and one LGMD2C). There was a statistically significant positive correlation between reduction of α-sarcoglycan level and disease severity in LGMD2D. Thirty-five mutations were identified in SGCA, SGCB, SGCG, and PMP22, 16 of which were novel. Exon 3 of SGCA was a hotspot region for mutations in LGMD2D. The missense mutation c.662G > A (p.R221H) was the most common mutation in SGCA. Missense mutations in both alleles of SGCA were associated with a relative benign disease course. No obvious clinical, sarcoglycan expression, and genetic correlation was found in LGMD2E. Conclusions This study expands the clinical and genetic spectrum of sarcoglycanopathies in Chinese patients and provides evidence that disease severity of LGMD2D may be predicted by α-sarcoglycan expression and SGCA mutation. Electronic supplementary material The online version of this article (10.1186/s13023-019-1021-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Zhiying Xie
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Yue Hou
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Yilin Liu
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Yanbin Fan
- Department of Pediatrics, Peking University First Hospital, Xishiku St, Xicheng District, Beijing, 100034, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China
| | - Hui Xiong
- Department of Pediatrics, Peking University First Hospital, Xishiku St, Xicheng District, Beijing, 100034, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, 8 Xishiku St, Xicheng District, Beijing, 100034, China.
| |
Collapse
|
17
|
Bulakh MV, Galeeva NM, Polyakova DA, Ryzhkova OP, Dadali EL, Polyakov AV. Molecular and Genetic Study of Limb-Girdle Muscular Dystrophy 2D in Patient Cohorts with Various Forms of Progressive Muscular Dystrophies. RUSS J GENET+ 2019. [DOI: 10.1134/s1022795419020030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
18
|
Carotti M, Marsolier J, Soardi M, Bianchini E, Gomiero C, Fecchio C, Henriques SF, Betto R, Sacchetto R, Richard I, Sandonà D. Repairing folding-defective α-sarcoglycan mutants by CFTR correctors, a potential therapy for limb-girdle muscular dystrophy 2D. Hum Mol Genet 2019; 27:969-984. [PMID: 29351619 PMCID: PMC5886177 DOI: 10.1093/hmg/ddy013] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 12/30/2017] [Indexed: 11/22/2022] Open
Abstract
Limb-girdle muscular dystrophy type 2D (LGMD2D) is a rare autosomal-recessive disease, affecting striated muscle, due to mutation of SGCA, the gene coding for α-sarcoglycan. Nowadays, more than 50 different SGCA missense mutations have been reported. They are supposed to impact folding and trafficking of α-sarcoglycan because the defective polypeptide, although potentially functional, is recognized and disposed of by the quality control of the cell. The secondary reduction of α-sarcoglycan partners, β-, γ- and δ-sarcoglycan, disrupts a key membrane complex that, associated to dystrophin, contributes to assure sarcolemma stability during muscle contraction. The complex deficiency is responsible for muscle wasting and the development of a severe form of dystrophy. Here, we show that the application of small molecules developed to rescue ΔF508-CFTR trafficking, and known as CFTR correctors, also improved the maturation of several α-sarcoglycan mutants that were consequently rescued at the plasma membrane. Remarkably, in myotubes from a patient with LGMD2D, treatment with CFTR correctors induced the proper re-localization of the whole sarcoglycan complex, with a consequent reduction of sarcolemma fragility. Although the mechanism of action of CFTR correctors on defective α-sarcoglycan needs further investigation, this is the first report showing a quantitative and functional recovery of the sarcoglycan-complex in human pathologic samples, upon small molecule treatment. It represents the proof of principle of a pharmacological strategy that acts on the sarcoglycan maturation process and we believe it has a great potential to develop as a cure for most of the patients with LGMD2D.
Collapse
Affiliation(s)
- Marcello Carotti
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Justine Marsolier
- Genethon, Evry F-91002, France.,INSERM, U951, INTEGRARE Research Unit, Evry F-91002, France
| | - Michela Soardi
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Elisa Bianchini
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy.,Aptuit, 37135 Verona, Italy
| | - Chiara Gomiero
- Department of Comparative Biomedicine and Food Science, University of Padova, Agripolis, 35020 Legnaro, Padova, Italy
| | - Chiara Fecchio
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| | - Sara F Henriques
- Genethon, Evry F-91002, France.,INSERM, U951, INTEGRARE Research Unit, Evry F-91002, France
| | - Romeo Betto
- Neuroscience Institute (CNR Padova), 35131 Padova, Italy
| | - Roberta Sacchetto
- Department of Comparative Biomedicine and Food Science, University of Padova, Agripolis, 35020 Legnaro, Padova, Italy
| | - Isabelle Richard
- Genethon, Evry F-91002, France.,INSERM, U951, INTEGRARE Research Unit, Evry F-91002, France
| | - Dorianna Sandonà
- Department of Biomedical Sciences, University of Padova, 35131 Padova, Italy
| |
Collapse
|
19
|
Cantero D, Hernández-Laín A, Martínez JFG, Pérez MR, Ruano Y, Lleixà C, Gallardo E, Domínguez-González C. Milder forms of α-sarcoglicanopathies diagnosed in adulthood by NGS analysis. J Neurol Sci 2018; 394:63-67. [DOI: 10.1016/j.jns.2018.08.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2018] [Revised: 08/21/2018] [Accepted: 08/27/2018] [Indexed: 10/28/2022]
|
20
|
Sarcoglycan Alpha Mitigates Neuromuscular Junction Decline in Aged Mice by Stabilizing LRP4. J Neurosci 2018; 38:8860-8873. [PMID: 30171091 DOI: 10.1523/jneurosci.0860-18.2018] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2018] [Revised: 08/07/2018] [Accepted: 08/22/2018] [Indexed: 01/08/2023] Open
Abstract
During aging, acetylcholine receptor (AChR) clusters become fragmented and denervated at the neuromuscular junction (NMJ). Underpinning molecular mechanisms are not well understood. We showed that LRP4, a receptor for agrin and critical for NMJ formation and maintenance, was reduced at protein level in aged mice, which was associated with decreased MuSK tyrosine phosphorylation, suggesting compromised agrin-LRP4-MuSK signaling in aged muscles. Transgenic expression of LRP4 in muscles alleviated AChR fragmentation and denervation and improved neuromuscular transmission in aged mice. LRP4 ubiquitination was augmented in aged muscles, suggesting increased LRP4 degradation as a mechanism for reduced LRP4. We found that sarcoglycan α (SGα) interacted with LRP4 and delayed LRP4 degradation in cotransfected cells. AAV9-mediated expression of SGα in muscles mitigated AChR fragmentation and denervation and improved neuromuscular transmission in aged mice. These observations support a model where compromised agrin-LRP4-MuSK signaling serves as a pathological mechanism of age-related NMJ decline and identify a novel function of SGα in stabilizing LRP4 for NMJ stability in aged mice.SIGNIFICANCE STATEMENT This study provides evidence that LRP4, a receptor of agrin that is critical for NMJ formation and maintenance, is reduced at protein level in aged muscles. Transgenic expression of LRP4 in muscles ameliorates AChR fragmentation and denervation and improves neuromuscular transmission in aged mice, demonstrating a critical role of the agrin-LRP4-MuSK signaling. Our study also reveals a novel function of SGα to prevent LRP4 degradation in aged muscles. Finally, we show that NMJ decline in aged mice can be mitigated by AAV9-mediated expression of SGα in muscles. These observations provide insight into pathological mechanisms of age-related NMJ decline and suggest that improved agrin-LRP4-MuSK signaling may be a target for potential therapeutic intervention.
Collapse
|
21
|
Bulakh MV, Ryzhkova OP, Polyakov AV. Sarcoglycanopathies: Clinical, Molecular and Genetic Characteristics, Epidemiology, Diagnostics and Treatment Options. RUSS J GENET+ 2018. [DOI: 10.1134/s1022795418020059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
22
|
Next-Generation Sequencing to Diagnose Muscular Dystrophy, Rhabdomyolysis, and HyperCKemia. Can J Neurol Sci 2018; 45:262-268. [DOI: 10.1017/cjn.2017.286] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
AbstractBackground:Neuromuscular disorders are a phenotypically and genotypically diverse group of diseases that can be difficult to diagnose accurately because of overlapping clinical features and nonspecific muscle pathology. Next-generation sequencing (NGS) is a high-throughput technology that can be used as a more time- and cost-effective tool for identifying molecular diagnoses for complex genetic conditions, such as neuromuscular disorders.Methods:One hundred and sixty-nine patients referred to a Canadian neuromuscular clinic for evaluation of possible muscle disease were screened with an NGS panel of muscular dystrophy–associated genes. Patients were categorized by the reason of referral (1) muscle weakness (n=135), (2) recurrent episodes of rhabdomyolysis (n=18), or (3) idiopathic hyperCKemia (n=16).Results:Pathogenic and likely pathogenic variants were identified in 36.09% of patients (61/169). The detection rate was 37.04% (50/135) in patients with muscle weakness, 33.33% (6/18) with rhabdomyolysis, and 31.25% (5/16) in those with idiopathic hyperCKemia.Conclusions:This study shows that NGS can be a useful tool in the molecular workup of patients seen in a neuromuscular clinic. Evaluating the utility of large panels of a muscle disease-specific NGS panel to investigate the genetic susceptibilities of rhabdomyolysis and/or idiopathic hyperCKemia is a relatively new field. Twenty-eight of the pathogenic and likely pathogenic variants reported here are novel and have not previously been associated with disease.
Collapse
|
23
|
Alavi A, Esmaeili S, Nilipour Y, Nafissi S, Tonekaboni SH, Zamani G, Ashrafi MR, Kahrizi K, Najmabadi H, Jazayeri F. LGMD2E is the most common type of sarcoglycanopathies in the Iranian population. J Neurogenet 2017; 31:161-169. [DOI: 10.1080/01677063.2017.1346093] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Afagh Alavi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Sara Esmaeili
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Yalda Nilipour
- Pediatric Pathology Research Center, Mofid Children Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahriar Nafissi
- Department of Neurology, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Hasan Tonekaboni
- Pediatric Neurology Center of Excellence, Department of Pediatric Neurology, Mofid Children Hospital, Faculty of Medicine, Shahid Beheshti Medical university, Tehran, Iran
| | - Gholamreza Zamani
- Children's Medical Center, Tehran University of Medical Sciences, Iranian Epilepsy Association Board, Tehran, Iran
| | - Mahmoud Reza Ashrafi
- Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Kimia Kahrizi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Hossein Najmabadi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Fatemeh Jazayeri
- Department of Neurology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
24
|
|
25
|
Turan S, Farruggio AP, Srifa W, Day JW, Calos MP. Precise Correction of Disease Mutations in Induced Pluripotent Stem Cells Derived From Patients With Limb Girdle Muscular Dystrophy. MOLECULAR THERAPY : THE JOURNAL OF THE AMERICAN SOCIETY OF GENE THERAPY 2016. [PMID: 26916285 DOI: 10.1038/mt.2016.40.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Limb girdle muscular dystrophies types 2B (LGMD2B) and 2D (LGMD2D) are degenerative muscle diseases caused by mutations in the dysferlin and alpha-sarcoglycan genes, respectively. Using patient-derived induced pluripotent stem cells (iPSC), we corrected the dysferlin nonsense mutation c.5713C>T; p.R1905X and the most common alpha-sarcoglycan mutation, missense c.229C>T; p.R77C, by single-stranded oligonucleotide-mediated gene editing, using the CRISPR/Cas9 gene-editing system to enhance the frequency of homology-directed repair. We demonstrated seamless, allele-specific correction at efficiencies of 0.7-1.5%. As an alternative, we also carried out precise gene addition strategies for correction of the LGMD2B iPSC by integration of wild-type dysferlin cDNA into the H11 safe harbor locus on chromosome 22, using dual integrase cassette exchange (DICE) or TALEN-assisted homologous recombination for insertion precise (THRIP). These methods employed TALENs and homologous recombination, and DICE also utilized site-specific recombinases. With DICE and THRIP, we obtained targeting efficiencies after selection of ~20%. We purified iPSC corrected by all methods and verified rescue of appropriate levels of dysferlin and alpha-sarcoglycan protein expression and correct localization, as shown by immunoblot and immunocytochemistry. In summary, we demonstrate for the first time precise correction of LGMD iPSC and validation of expression, opening the possibility of cell therapy utilizing these corrected iPSC.
Collapse
Affiliation(s)
- Soeren Turan
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Alfonso P Farruggio
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Waracharee Srifa
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - John W Day
- Department of Neurology, Stanford University School of Medicine, Stanford, California, USA
| | - Michele P Calos
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
26
|
Turan S, Farruggio AP, Srifa W, Day JW, Calos MP. Precise Correction of Disease Mutations in Induced Pluripotent Stem Cells Derived From Patients With Limb Girdle Muscular Dystrophy. Mol Ther 2016; 24:685-96. [PMID: 26916285 DOI: 10.1038/mt.2016.40] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 02/09/2016] [Indexed: 12/22/2022] Open
Abstract
Limb girdle muscular dystrophies types 2B (LGMD2B) and 2D (LGMD2D) are degenerative muscle diseases caused by mutations in the dysferlin and alpha-sarcoglycan genes, respectively. Using patient-derived induced pluripotent stem cells (iPSC), we corrected the dysferlin nonsense mutation c.5713C>T; p.R1905X and the most common alpha-sarcoglycan mutation, missense c.229C>T; p.R77C, by single-stranded oligonucleotide-mediated gene editing, using the CRISPR/Cas9 gene-editing system to enhance the frequency of homology-directed repair. We demonstrated seamless, allele-specific correction at efficiencies of 0.7-1.5%. As an alternative, we also carried out precise gene addition strategies for correction of the LGMD2B iPSC by integration of wild-type dysferlin cDNA into the H11 safe harbor locus on chromosome 22, using dual integrase cassette exchange (DICE) or TALEN-assisted homologous recombination for insertion precise (THRIP). These methods employed TALENs and homologous recombination, and DICE also utilized site-specific recombinases. With DICE and THRIP, we obtained targeting efficiencies after selection of ~20%. We purified iPSC corrected by all methods and verified rescue of appropriate levels of dysferlin and alpha-sarcoglycan protein expression and correct localization, as shown by immunoblot and immunocytochemistry. In summary, we demonstrate for the first time precise correction of LGMD iPSC and validation of expression, opening the possibility of cell therapy utilizing these corrected iPSC.
Collapse
Affiliation(s)
- Soeren Turan
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Alfonso P Farruggio
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Waracharee Srifa
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - John W Day
- Department of Neurology, Stanford University School of Medicine, Stanford, California, USA
| | - Michele P Calos
- Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
27
|
Liang WC, Chou PC, Hung CC, Su YN, Kan TM, Chen WZ, Hayashi YK, Nishino I, Jong YJ. Probable high prevalence of limb-girdle muscular dystrophy type 2D in Taiwan. J Neurol Sci 2016; 362:304-8. [PMID: 26944168 DOI: 10.1016/j.jns.2016.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 01/14/2016] [Accepted: 02/01/2016] [Indexed: 01/05/2023]
Abstract
Limb-girdle muscular dystrophy type 2D (LGMD2D), an autosomal-recessive inherited LGMD, is caused by the mutations in SGCA. SGCA encodes alpha-sarcoglycan (SG) that forms a heterotetramer with other SGs in the sarcolemma, and comprises part of the dystrophin-glycoprotein complex. The frequency of LGMD2D is variable among different ethnic backgrounds, and so far only a few patients have been reported in Asia. We identified five patients with a novel homozygous mutation of c.101G>T (p.Arg34Leu) in SGCA from a big aboriginal family ethnically consisting of two tribes in Taiwan. Patient 3 is the maternal uncle of patients 1 and 2. All their parents, heterozygous for c.101G>T, denied consanguineous marriages although they were from the same tribe. The heterozygous parents of patients 4 and 5 were from two different tribes, originally residing in different geographic regions in Taiwan. Haplotype analysis showed that all five patients shared the same mutation-associated haplotype, indicating the probability of a founder effect and consanguinity. The results suggest that the carrier rate of c.101G>T in SGCA may be high in Taiwan, especially in the aboriginal population regardless of the tribes. It is important to investigate the prevalence of LGMD2D in Taiwan for early diagnosis and treatment.
Collapse
Affiliation(s)
- Wen-Chen Liang
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Po-Ching Chou
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | | | - Yi-Ning Su
- Sofiva Genomics Co., Ltd., Taipei, Taiwan; Dianthus Maternal Fetal Medicine Clinic, Taipei, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tsu-Min Kan
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wan-Zi Chen
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yukiko K Hayashi
- Department of Neurophysiology, Tokyo Medical University, Tokyo, Japan; Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan; Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yuh-Jyh Jong
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan; Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan; Department of Biological Science and Technology, Institute of Molecular Medicine and Bioengineering, College of Biological Science and Technology, National Chiao Tung University, Hsinchu, Taiwan.
| |
Collapse
|
28
|
Tarnopolsky M, Hoffman E, Giri M, Shoffner J, Brady L. Alpha-sarcoglycanopathy presenting as exercise intolerance and rhabdomyolysis in two adults. Neuromuscul Disord 2015; 25:952-4. [DOI: 10.1016/j.nmd.2015.09.010] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 09/09/2015] [Accepted: 09/14/2015] [Indexed: 11/28/2022]
|
29
|
Townsend D. Finding the sweet spot: assembly and glycosylation of the dystrophin-associated glycoprotein complex. Anat Rec (Hoboken) 2014; 297:1694-705. [PMID: 25125182 PMCID: PMC4135523 DOI: 10.1002/ar.22974] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 03/27/2014] [Indexed: 01/12/2023]
Abstract
The dystrophin-associated glycoprotein complex (DGC) is a collection of glycoproteins that are essential for the normal function of striated muscle and many other tissues. Recent genetic studies have implicated the components of this complex in over a dozen forms of muscular dystrophy. Furthermore, disruption of the DGC has been implicated in many forms of acquired disease. This review aims to summarize the current state of knowledge regarding the processing and assembly of dystrophin-associated proteins with a focus primarily on the dystroglycan heterodimer and the sarcoglycan complex. These proteins form the transmembrane portion of the DGC and undergo a complex multi-step processing with proteolytic cleavage, differential assembly, and both N- and O-glycosylation. The enzymes responsible for this processing and a model describing the sequence and subcellular localization of these events are discussed.
Collapse
Affiliation(s)
- Dewayne Townsend
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota
| |
Collapse
|
30
|
Concomitant alpha- and gamma-sarcoglycan deficiencies in a Turkish boy with a novel deletion in the alpha-sarcoglycan gene. Case Rep Genet 2014; 2014:248561. [PMID: 25050186 PMCID: PMC4090428 DOI: 10.1155/2014/248561] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Accepted: 06/02/2014] [Indexed: 11/19/2022] Open
Abstract
Limb-girdle muscular dystrophy type 2D (LGMD-2D) is caused by autosomal recessive defects in the alpha-sarcoglycan gene located on chromosome 17q21. In this study, we present a child with alpha-sarcoglycanopathy and describe a novel deletion in the alpha-sarcoglycan gene. A 5-year-old boy had a very high serum creatinine phosphokinase level, which was determined incidentally, and a negative molecular test for the dystrophin gene. Muscle biopsy showed dystrophic features. Immunohistochemistry showed that there was diminished expression of alpha- and gamma-sarcoglycans. DNA analysis revealed a novel 7 bp homozygous deletion in exon 3 of the alpha-sarcoglycan gene. His parents were consanguineous heterozygous carriers of the same deletion. We believe this is the first confirmed case of primary alpha-sarcoglycanopathy with a novel deletion in Turkey. In addition, this study demonstrated that both muscle biopsy and DNA analysis remain important methods for the differential diagnosis of muscular dystrophies because dystrophinopathies and sarcoglycanopathies are so similar.
Collapse
|
31
|
Diniz G, Tosun Yildirim H, Akinci G, Hazan F, Ozturk A, Yararbas K, Tukun A. Sarcolemmal alpha and gamma sarcoglycan protein deficiencies in Turkish siblings with a novel missense mutation in the alpha sarcoglycan gene. Pediatr Neurol 2014; 50:640-7. [PMID: 24742800 DOI: 10.1016/j.pediatrneurol.2013.12.024] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 12/08/2013] [Accepted: 12/11/2013] [Indexed: 11/26/2022]
Abstract
BACKGROUND The sarcoglycan alpha gene, also known as the adhalin gene, is located on chromosome 17q21; mutations in this gene are associated with limb-girdle muscular dystrophy type 2D. We describe two Turkish siblings with findings consistent with limb-girdle muscular dystrophy type 2D. The evaluation excluded a dystrophinopathy, which is the most common form of muscular dystrophy. PATIENTS Both siblings had very high levels of creatinine phosphokinase and negative molecular tests for deletions and duplications of the dystrophin gene. The older boy presented at 8 years of age with an inability to climb steps and an abnormal gait. His younger brother was 5 years old and had similar symptoms. The muscle biopsy evaluation was performed only in the older brother. RESULTS The muscle biopsy showed dystrophic features as well as a deficiency in the expression of two different glycoproteins: the alpha sarcoglycan and the gamma sarcoglycan. Sarcolemmal expressions of dystrophin and other sarcoglycans (beta and delta) were diffusely present. DNA analysis demonstrated the presence of previously unknown homozygous mutations [c.226 C > T (p.L76 F)] in exon 3 in the sarcoglycan alpha genes of both siblings. Similar heterozygous point mutations at the same locus were found in both parents, but the genes of beta, delta, and gamma sarcoglycan were normal in the remaining family members. CONCLUSIONS We describe two siblings with limb-girdle muscular dystrophy type 2D with a novel missense mutation. These patients illustrate that the differential diagnosis of muscular dystrophies is impossible with clinical findings alone. Therefore, a muscle biopsy and DNA analysis remain essential methods for diagnosis of muscle diseases.
Collapse
Affiliation(s)
- Gulden Diniz
- Basic Oncology, Associate Professor in Pathology, Tepecik Research Hospital, Izmir, Turkey.
| | - Hulya Tosun Yildirim
- Pathology Department, Dr. Behcet Uz Children's, Research Hospital, Izmir, Turkey
| | - Gulcin Akinci
- Pediatric Neurology Department, Dr. Behcet Uz Children's, Research Hospital, Izmir, Turkey
| | - Filiz Hazan
- Medical Genetics Department, Dr. Behcet Uz Children's, Research Hospital, Izmir, Turkey
| | - Aysel Ozturk
- Pediatric Neurology Department, Dr. Behcet Uz Children's, Research Hospital, Izmir, Turkey
| | - Kanay Yararbas
- Medical Genetics Department, Duzen Laboratories, Istanbul and Ankara, Turkey
| | - Ajlan Tukun
- Medical Genetics Department, Duzen Laboratories, Istanbul and Ankara, Turkey; Medical Genetics Department, Ankara University Medical School, Ankara, Turkey
| |
Collapse
|
32
|
Thin, a Trim32 ortholog, is essential for myofibril stability and is required for the integrity of the costamere in Drosophila. Proc Natl Acad Sci U S A 2012; 109:17983-8. [PMID: 23071324 DOI: 10.1073/pnas.1208408109] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Myofibril stability is required for normal muscle function and maintenance. Mutations that disrupt myofibril stability result in individuals who develop progressive muscle wasting, or muscular dystrophy, and premature mortality. Here we present our investigations of the Drosophila l(2)thin [l(2)tn] mutant. The "thin" phenotype exhibits features of the human muscular disease phenotype in that tn mutant larvae show progressive muscular degeneration. Loss-of-function and rescue experiments determined that l(2)tn is allelic to the tn locus [previously annotated as both CG15105 and another b-box affiliate (abba)]. tn encodes a TRIM (tripartite motif) containing protein highly expressed in skeletal muscle and is orthologous to the human limb-girdle muscular dystrophy type 2H disease gene Trim32. Thin protein is localized at the Z-disk in muscle, but l(2)tn mutants showed no genetic interaction with mutants affecting the Z-line-associated protein muscle LIM protein 84B. l(2)tn, along with loss-of-function mutants generated for tn, showed no relative mislocalization of the Z-disk proteins α-Actinin and muscle LIM protein 84B. In contrast, tn mutants had significant disorganization of the costameric orthologs β-integrin, Spectrin, Talin, and Vinculin, and we present the initial description for the costamere, a key muscle stability complex, in Drosophila. Our studies demonstrate that myofibrils progressively unbundle in flies that lack Thin function through progressive costamere breakdown. Due to the high conservation of these structures in animals, we demonstrate a previously unknown role for TRIM32 proteins in myofibril stability.
Collapse
|
33
|
Soheili T, Gicquel E, Poupiot J, N'Guyen L, Le Roy F, Bartoli M, Richard I. Rescue of sarcoglycan mutations by inhibition of endoplasmic reticulum quality control is associated with minimal structural modifications. Hum Mutat 2011; 33:429-39. [DOI: 10.1002/humu.21659] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2011] [Accepted: 10/31/2011] [Indexed: 11/08/2022]
|
34
|
Abstract
The so-called sarcoglycanopathies form a subgroup of four genetically closely related autosomal recessive limb-girdle muscular dystrophies (LGMD2C-F) caused by mutations of the α-, β-, γ-, and δ-sarcoglycan genes. All four sarcoglycans are glycosylated transmembrane proteins and form a tetrameric complex that is part of dystrophin-associated proteins. The clinical phenotype associated with sarcoglycanopathies is characterized by a slowly progressive proximal muscle weakness with onset during childhood in most cases. The disease course is often similar but more variable than X-linked Duchenne muscular dystrophy. Diagnosis is usually based on muscle biopsy findings that confirm dystrophic changes and deficiency of one or more sarcoglycan proteins. Genetic testing is used to confirm the diagnosis. A number of different animal models have been developed to study the function of sarcoglycans and to develop specific therapeutic strategies such as gene transfer, but so far none of these techniques has entered clinical practice. Therefore, treatment is symptomatic and aims at amelioration of locomotor, respiratory, and cardiac manifestations of the disease.
Collapse
Affiliation(s)
- Janbernd Kirschner
- Division of Neuropediatrics and Muscle Disorders, University Medical Center Freiburg, Freiburg, Germany.
| | | |
Collapse
|
35
|
Mendell JR, Rodino-Klapac LR, Rosales XQ, Coley BD, Galloway G, Lewis S, Malik V, Shilling C, Byrne BJ, Conlon T, Campbell KJ, Bremer WG, Taylor LE, Flanigan KM, Gastier-Foster JM, Astbury C, Kota J, Sahenk Z, Walker CM, Clark KR. Sustained alpha-sarcoglycan gene expression after gene transfer in limb-girdle muscular dystrophy, type 2D. Ann Neurol 2010; 68:629-38. [PMID: 21031578 PMCID: PMC2970162 DOI: 10.1002/ana.22251] [Citation(s) in RCA: 175] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECTIVE The aim of this study was to attain long-lasting alpha-sarcoglycan gene expression in limb-girdle muscular dystrophy, type 2D (LGMD2D) subjects mediated by adeno-associated virus (AAV) gene transfer under control of a muscle specific promoter (tMCK). METHODS rAAV1.tMCK.hSGCA (3.25 × 10¹¹ vector genomes) was delivered to the extensor digitorum brevis muscle of 3 subjects with documented SGCA mutations via a double-blind, randomized, placebo controlled trial. Control sides received saline. The blind was not broken until the study was completed at 6 months and all results were reported to the oversight committee. RESULTS Persistent alpha-sarcoglycan gene expression was achieved for 6 months in 2 of 3 LGMD2D subjects. Markers for muscle fiber transduction other than alpha-sarcoglycan included expression of major histocompatibility complex I, increase in muscle fiber size, and restoration of the full sarcoglycan complex. Mononuclear inflammatory cells recruited to the site of gene transfer appeared to undergo programmed cell death, demonstrated by terminal deoxynucleotide transferase-mediated deoxyuridine triphosphate nick-end labeling and caspase-3 staining. A patient failing gene transfer demonstrated an early rise in neutralizing antibody titers and T-cell immunity to AAV, validated by enzyme-linked immunospot on the second day after gene injection. This was in clear distinction to other participants with satisfactory gene expression. INTERPRETATION The findings of this gene replacement study in LGMD2D subjects have important implications not previously demonstrated in muscular dystrophy. Long-term, sustainable gene expression of alpha-sarcoglycan was observed following gene transfer mediated by AAV. The merit of a muscle-specific tMCK promoter, not previously used in a clinical trial, was evident, and the potential for reversal of disease was displayed.
Collapse
Affiliation(s)
- Jerry R Mendell
- Department of Pediatrics, Ohio State University, Columbus, OH, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
|
37
|
Reverse protein arrays as novel approach for protein quantification in muscular dystrophies. Neuromuscul Disord 2010; 20:302-9. [PMID: 20304647 DOI: 10.1016/j.nmd.2010.02.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2009] [Revised: 02/19/2010] [Accepted: 02/23/2010] [Indexed: 11/23/2022]
Abstract
The definite molecular diagnosis in patients with muscular dystrophies often requires the assessment of muscular expression of multiple proteins in small amounts of muscle tissue. The sample material obtained in muscle biopsies is limited and the measurement of multiple proteins is often restricted to conventional, non-quantitative assays, i.e. immunohistochemistry and immunoblotting. Here, we demonstrate that reverse protein arrays are a novel and excellent material-saving method for the measurement and quantification of changes in protein expression between healthy and diseased muscle tissue as well as cultured primary myotubes. We evaluated a set of antibodies and found reproducible differences between Duchenne muscular dystrophy/limb-girdle muscular dystrophy patients and control samples for dystrophin, the sarcoglycans and the dystroglycans. As little as 10 mg of tissue is sufficient for the analysis of all diagnostically relevant proteins. The average coefficient of variation calculated for the sample signals confirmed that the method is highly reproducible. Thus, our experiments provide strong evidence that quantitative protein detection from very small amounts of muscle tissue is possible using reverse protein arrays. This technology may not only be of interest for diagnostic purposes, but also for protein quantification of multiple, follow-up biopsies during clinical trials when protein expression in muscle is considered an important outcome measure or biomarker.
Collapse
|
38
|
Navarro C, Teijeira S. Molecular diagnosis of muscular dystrophies, focused on limb girdle muscular dystrophies. ACTA ACUST UNITED AC 2009; 3:631-47. [PMID: 23496048 DOI: 10.1517/17530050903313988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Muscular dystrophies include a spectrum of muscle disorders, some of which are phenotypically well characterized. The identification of dystrophin as the causative factor in Duchenne muscular dystrophy has led to the development of molecular genetics and has facilitated the division of muscular dystrophies into distinct groups, among which are the 'limb girdle muscular dystrophies'. OBJECTIVES This article reviews the methodology to be used in the diagnosis of muscular dystrophies, focused on the groups of limb girdle muscular dystrophies, and the development of new strategies to reach a final molecular diagnosis. METHOD A literature review (Medline) from 1985 to the present. CONCLUSION Immunohistochemistry and western blotting analyses of the proteins involved in the various forms of muscular dystrophies have permitted a refined pathological approach necessary to conduct genetic studies and to offer appropriate genetic counseling. The application of molecular medicine in genetic muscular dystrophies also brings great hope to the therapeutic management of these patients.
Collapse
Affiliation(s)
- Carmen Navarro
- University Hospital of Vigo, Department of Pathology and Neuropathology, Meixoeiro, s/n, 36200 Vigo - Pontevedra, Spain +34 986 81 11 11 ext. 211661 ; +34 986 27 64 16 ;
| | | |
Collapse
|
39
|
Mendell JR, Rodino-Klapac LR, Rosales-Quintero X, Kota J, Coley BD, Galloway G, Craenen JM, Lewis S, Malik V, Shilling C, Byrne BJ, Conlon T, Campbell KJ, Bremer WG, Viollet L, Walker CM, Sahenk Z, Clark KR. Limb-girdle muscular dystrophy type 2D gene therapy restores alpha-sarcoglycan and associated proteins. Ann Neurol 2009; 66:290-7. [PMID: 19798725 DOI: 10.1002/ana.21732] [Citation(s) in RCA: 158] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE alpha-Sarcoglycan deficiency results in a severe form of muscular dystrophy (limb-girdle muscular dystrophy type 2D [LGMD2D]) without treatment. Gene replacement represents a strategy for correcting the underlying defect. Questions related to this approach were addressed in this clinical trial, particularly the need for immunotherapy and persistence of gene expression. METHODS A double-blind, randomized controlled trial using rAAV1.tMCK.hSGCA injected into the extensor digitorum brevis muscle was conducted. Control sides received saline. A 3-day course of methylprednisolone accompanied gene transfer without further immune suppression. RESULTS No adverse events were encountered. SGCA gene expression increased 4-5-fold over control sides when examined at 6 weeks (2 subjects) and 3 months (1 subject). The full sarcoglycan complex was restored in all subjects, and muscle fiber size was increased in the 3-month subject. Adeno-associated virus serotype 1 (AAV1)-neutralizing antibodies were seen as early as 2 weeks. Neither CD4+ nor CD8+ cells were increased over contralateral sides. Scattered foci of inflammation could be found, but showed features of programmed cell death. Enzyme-linked immunospot (ELISpot) showed no interferon-gamma response to alpha-SG or AAV1 capsid peptide pools, with the exception of a minimal capsid response in 1 subject. Restimulation to detect low-frequency capsid-specific T cells by ELISpot assays was negative. Results of the first 3 subjects successfully achieved study aims, precluding the need for additional enrollment. INTERPRETATION The finding of this gene replacement study in LGMD2D has important implications for muscular dystrophy. Sustained gene expression was seen, but studies over longer time periods without immunotherapy will be required for design of vascular delivery gene therapy trials.
Collapse
Affiliation(s)
- Jerry R Mendell
- Department of Pediatrics, Ohio State University, Columbus, OH, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Sarcoglycanopathies are a group of autosomal recessive muscle-wasting disorders caused by genetic defects in one of four cell membrane glycoproteins, alpha-, beta-, gamma- or delta-sarcoglycan. These four sarcoglycans form a subcomplex that is closely linked to the major dystrophin-associated protein complex, which is essential for membrane integrity during muscle contraction and provides a scaffold for important signalling molecules. Proper assembly, trafficking and targeting of the sarcoglycan complex is of vital importance, and mutations that severely perturb tetramer formation and localisation result in sarcoglycanopathy. Gene defects in one sarcoglycan cause the absence or reduced concentration of the other subunits. Most genetic defects generate mutated proteins that are degraded through the cell's quality control system; however, in many cases, conformational modifications do not affect the function of the protein, yet it is recognised as misfolded and prematurely degraded. Recent evidence shows that misfolded sarcoglycans could be rescued to the cell membrane by assisting their maturation along the ER secretory pathway. This review summarises the etiopathogenesis of sarcoglycanopathies and highlights the quality control machinery as a potential pharmacological target for therapy of these genetic disorders.
Collapse
Affiliation(s)
- Dorianna Sandonà
- Department of Biomedical Sciences, University of Padova, 35121
Padova, Italy
| | - Romeo Betto
- C.N.R. Institute of Neuroscience, Neuromuscular Biology and
Physiopathology, 35121 Padova, Italy
| |
Collapse
|
41
|
|
42
|
Wildförster V, Dekomien G. Detecting copy number variations in autosomal recessive limb-girdle muscular dystrophies using a multiplex ligation-dependent probe amplification (MLPA) assay. Mol Cell Probes 2008; 23:55-9. [PMID: 19056483 DOI: 10.1016/j.mcp.2008.11.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2008] [Revised: 11/06/2008] [Accepted: 11/06/2008] [Indexed: 10/21/2022]
Abstract
Pathogenic mutations in the four sarcoglycan genes, designated SGCA, SGCB, SGCD and SGCG, are responsible for a subgroup of autosomal, recessive limb-girdle muscular dystrophies (LGMD 2C-F), also called sarcoglycanopathies. For the present study, we designed a multiplex ligation-dependent probe amplification (MLPA) assay, targeting all 30 coding exons and a non-coding exon of these four genes. The assay uses synthetic probes and two colours, such that as many as 28 probes can be combined into one reaction. The set of probes was established for routine application, in order to diagnostically screen patients for large duplications or deletions. In 14 of the 94 cases (15%) tested, we detected changes in copy number. Mutations in gene SGCG accounted for 7 of the 94 cases (8%), suggesting that the size of the gene makes it vulnerable to large exonic deletions. The results suggested that all cases of sarcoglycanopathy should be screened for changes in copy number. The MLPA was shown to be a rapid, robust and reliable method to screen for copy number variations (CNVs). The present synthetic probe-approach overcomes the limitations associated with cloning procedures and is particularly applicable to a range of diseases for which the number of patients to be tested is small.
Collapse
Affiliation(s)
- Verena Wildförster
- Human Genetics, Ruhr University Bochum, Universitaetsstrabe 150, 44801 Bochum, Germany
| | | |
Collapse
|
43
|
Davis J, Westfall MV, Townsend D, Blankinship M, Herron TJ, Guerrero-Serna G, Wang W, Devaney E, Metzger JM. Designing heart performance by gene transfer. Physiol Rev 2008; 88:1567-651. [PMID: 18923190 DOI: 10.1152/physrev.00039.2007] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The birth of molecular cardiology can be traced to the development and implementation of high-fidelity genetic approaches for manipulating the heart. Recombinant viral vector-based technology offers a highly effective approach to genetically engineer cardiac muscle in vitro and in vivo. This review highlights discoveries made in cardiac muscle physiology through the use of targeted viral-mediated genetic modification. Here the history of cardiac gene transfer technology and the strengths and limitations of viral and nonviral vectors for gene delivery are reviewed. A comprehensive account is given of the application of gene transfer technology for studying key cardiac muscle targets including Ca(2+) handling, the sarcomere, the cytoskeleton, and signaling molecules and their posttranslational modifications. The primary objective of this review is to provide a thorough analysis of gene transfer studies for understanding cardiac physiology in health and disease. By comparing results obtained from gene transfer with those obtained from transgenesis and biophysical and biochemical methodologies, this review provides a global view of cardiac structure-function with an eye towards future areas of research. The data presented here serve as a basis for discovery of new therapeutic targets for remediation of acquired and inherited cardiac diseases.
Collapse
Affiliation(s)
- Jennifer Davis
- Department of Integrative Biology and Physiology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Gastaldello S, D'Angelo S, Franzoso S, Fanin M, Angelini C, Betto R, Sandonà D. Inhibition of proteasome activity promotes the correct localization of disease-causing alpha-sarcoglycan mutants in HEK-293 cells constitutively expressing beta-, gamma-, and delta-sarcoglycan. THE AMERICAN JOURNAL OF PATHOLOGY 2008; 173:170-81. [PMID: 18535179 PMCID: PMC2438295 DOI: 10.2353/ajpath.2008.071146] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/27/2008] [Indexed: 11/20/2022]
Abstract
Sarcoglycanopathies are progressive muscle-wasting disorders caused by genetic defects of four proteins, alpha-, beta-, gamma-, and delta-sarcoglycan, which are elements of a key transmembrane complex of striated muscle. The proper assembly of the sarcoglycan complex represents a critical issue of sarcoglycanopathies, as several mutations severely perturb tetramer formation. Misfolded proteins are generally degraded through the cell's quality-control system; however, this can also lead to the removal of some functional polypeptides. To explore whether it is possible to rescue sarcoglycan mutants by preventing their degradation, we generated a heterologous cell system, based on human embryonic kidney (HEK) 293 cells, constitutively expressing three (beta, gamma, and delta) of the four sarcoglycans. In these betagammadelta-HEK cells, the lack of alpha-sarcoglycan prevented complex formation and cell surface localization, wheras the presence of alpha-sarcoglycan allowed maturation and targeting of the tetramer. As in muscles of sarcoglycanopathy patients, transfection of betagammadelta-HEK cells with disease-causing alpha-sarcoglycan mutants led to dramatic reduction of the mutated proteins and the absence of the complex from the cell surface. Proteasomal inhibition reduced the degradation of mutants and facilitated the assembly and targeting of the sarcoglycan complex to the plasma membrane. These data provide important insights for the potential development of pharmacological therapies for sarcoglycanopathies.
Collapse
|
45
|
Trabelsi M, Kavian N, Daoud F, Commere V, Deburgrave N, Beugnet C, Llense S, Barbot JC, Vasson A, Kaplan JC, Leturcq F, Chelly J. Revised spectrum of mutations in sarcoglycanopathies. Eur J Hum Genet 2008; 16:793-803. [PMID: 18285821 DOI: 10.1038/ejhg.2008.9] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
To define the spectrum of mutations in alpha-, beta-, gamma-, and delta-sarcoglycan (SG) genes, we analyzed these genes in 69 probands with clinical and biological criteria compatible with the diagnosis of autosomal recessive limb-girdle muscular dystrophy. For 48 patients, muscle biopsies were available and multiplex western blot analysis of muscle proteins showed significant abnormalities of alpha- and gamma-SG. Our diagnostic strategy includes multiplex western blot, sequencing of SG genes, multiplex quantitative-fluorescent PCR and RT-PCR analyses. Mutations were detected in 57 patients and homozygous or compound heterozygous mutations were identified in 75% (36/48) of the patients with abnormal western blot, and in 52% (11/21) of the patients without muscle biopsy. Involvement of alpha-SG was demonstrated in 55.3% of cases (26/47), whereas gamma- and beta-SG were implicated in 25.5% (12/47) and in 17% (8/47) of cases, respectively. Interestingly, we identified 25 novel mutations, and a significant proportion of these mutations correspond to deletions (identified in 14 patients) of complete exon(s) of alpha- or gamma-SG genes, and partial duplications (identified in 5 patients) of exon 1 of beta-SG gene. This study highlights the high frequency of exonic deletions of alpha- and gamma-SG genes, as well as the presence of a hotspot of duplications affecting exon 1 of the beta-SG gene. In addition, protein analysis by multiplex western blot in combination with mutation screening and genotyping results allowed to propose a comprehensive and efficient diagnostic strategy and strongly suggested the implication of additional genes, yet to be identified, in sarcoglycanopathy-like disorders.
Collapse
Affiliation(s)
- Madiha Trabelsi
- Laboratoire de Biochimie Génétique et Moléculaire, Hôpital Cochin, Paris, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Kobuke K, Piccolo F, Garringer KW, Moore SA, Sweezer E, Yang B, Campbell KP. A common disease-associated missense mutation in alpha-sarcoglycan fails to cause muscular dystrophy in mice. Hum Mol Genet 2008; 17:1201-13. [PMID: 18252746 DOI: 10.1093/hmg/ddn009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Limb-girdle muscular dystrophy type 2D (LGMD2D) is caused by autosomal recessive mutations in the alpha-sarcoglycan gene. An R77C substitution is the most prevalent cause of the disease, leading to disruption of the sarcoglycan-sarcospan complex. To model this common mutation, we generated knock-in mice with an H77C substitution in alpha-sarcoglycan. The floxed neomycin (Neo)-cassette retained at the targeted H77C alpha-sarcoglycan locus caused a loss of alpha-sarcoglycan expression, resulting in muscular dystrophy in homozygotes, whereas Cre-mediated deletion of the floxed Neo-cassette led to recovered H77C alpha-sarcoglycan expression. Contrary to expectations, mice homozygous for the H77C-encoding allele expressed both this mutant alpha-sarcoglycan and the other components of the sarcoglycan-sarcospan complex in striated muscle, and did not develop muscular dystrophy. Accordingly, conditional rescued expression of the H77C protein in striated muscle of the alpha-sarcoglycan-deficient mice prevented the disease. Adding to the case that the behavior of mutant alpha-sarcoglycan is different between humans and mice, mutant human R77C alpha-sarcoglycan restored the expression of the sarcoglycan-sarcospan complex when introduced by adenoviral vector into the skeletal muscle of previously created alpha-sarcoglycan null mice. These findings indicate that the alpha-sarcoglycan with the most frequent missense mutation in LGMD2D is correctly processed, is transported to the sarcolemma, and is fully functional in mouse muscle. Our study presents an unexpected difference in the behavior of a missense-mutated protein in mice versus human patients, and emphasizes the need to understand species-specific protein quality control systems.
Collapse
Affiliation(s)
- Kazuhiro Kobuke
- Howard Hughes Medical Institute, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | |
Collapse
|
47
|
Bartoli M, Gicquel E, Barrault L, Soheili T, Malissen M, Malissen B, Vincent-Lacaze N, Perez N, Udd B, Danos O, Richard I. Mannosidase I inhibition rescues the human α-sarcoglycan R77C recurrent mutation. Hum Mol Genet 2008; 17:1214-21. [PMID: 18252745 DOI: 10.1093/hmg/ddn029] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|
48
|
Draviam RA, Wang B, Shand SH, Xiao X, Watkins SC. α-Sarcoglycan is Recycled from the Plasma Membrane in the Absence of Sarcoglycan Complex Assembly. Traffic 2006; 7:793-810. [PMID: 16787395 DOI: 10.1111/j.1600-0854.2006.00433.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The sarcoglycan complex consists of four subunits in skeletal muscle (alpha, beta, gamma, and delta-SG). Mutations in alpha-sarcoglycan (alpha-SG) result in the most common form of limb girdle muscular dystrophy. However, the function of alpha-SG remains unknown. In this report we attempt to clarify its function by delineating the trafficking pathway of alpha-SG in live cells. We present evidence, utilizing total internal reflection microscopy, fluorescence recovery after photobleaching and photoactivation of green fluorescent protein (GFP) constructs, that pools of alpha-SG are able to translocate to the plasma membrane in the absence of the remaining sarcoglycans. Internalization assays and drug treatment experiments demonstrate that alpha-SG recycles from the plasma membrane and accumulates in recycling endosomes. We also establish that alpha-SG utilizes well-described clathrin mediated mechanisms and microtubules to traffic within the cell. Finally, we show that the most commonly reoccurring limb girdle muscular dystrophy (R77C) mutation causes a fundamental defect in protein biosynthesis, trapping the mutant protein in the endoplasmic recticulum (ER). These results demonstrate that alpha-SG requires assembly into the sarcoglycan complex for stability at the plasma membrane rather than export out of the ER. Furthermore, this data suggests that alpha-SG utilizes known trafficking machinery to control deposition at the plasma membrane through recycling.
Collapse
Affiliation(s)
- Romesh A Draviam
- Department of Cell biology and Molecular Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261, USA
| | | | | | | | | |
Collapse
|
49
|
Hackman P, Juvonen V, Sarparanta J, Penttinen M, Aärimaa T, Uusitalo M, Auranen M, Pihko H, Alén R, Junes M, Lönnqvist T, Kalimo H, Udd B. Enrichment of the R77C α-sarcoglycan gene mutation in finnish LGMD2D patients. Muscle Nerve 2004; 31:199-204. [PMID: 15736300 DOI: 10.1002/mus.20267] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Limb-girdle muscular dystrophy 2D (LGMD2D) is caused by mutations in the alpha-sarcoglycan gene (SGCA). The most frequently reported mutation, 229CGC>TGC (R77C) in exon 3 of SGCA, results in the substitution of arginine by cysteine. We present here the clinical, immunohistochemical, and genetic data of 11 Finnish patients with LGMD2D caused by mutations in SGCA. Mutational analysis showed 10 patients homozygous and 1 compound heterozygous for R77C. A wide spectrum of SGCA mutations has been reported previously. Our results show an enrichment of R77C in Finland, further underlined by the observed carrier frequency of 1 per 150. According to the annual birth rate of approximately 60,000 in Finland, one LGMD2D patient with a homozygous mutation is expected to be born every 1 or 2 years on average. The presence of an ancient founder mutation is indicated by the fact that all patients shared a short common haplotype extending > or = 790 kilobases. Our results emphasize the need to include the SGCA gene R77C mutation test in routine DNA analyses of severe dystrophinopathy-like muscular dystrophies in Finland, and suggest that the applicability of this test in other populations should be studied as well.
Collapse
Affiliation(s)
- P Hackman
- Folkhälsan Institute of Genetics and Department of Medical Genetics, University of Helsinki, Biomedicum, Helsinki, Finland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Limb girdle muscular dystrophies (LGMDs) are a genetically heterogeneous group of primary myopathies involving progressive weakness and wasting of the muscles in the hip and shoulder girdles, with distal spread to the bulbar or respiratory musculature in rare cases. Depending on the mode of genetic transmission, six autosomal dominant forms (LGMD1A-F, 10-25%) and ten autosomal recessive forms (LGMD2A-J, 75-90%) are currently known. The prevalence of LGMDs is 0.8/100,000. These conditions are caused by mutations in genes encoding for myotilin (5q31, LGMD1A), lamin A/C (1q11-q21.2, LGMD1B), caveolin-3 (3p25, LGMD1C), unknown proteins (7q, LGMD1D, 6q23, LGMD1E, 7q32.1-32.2., LGMD1F), calpain-3 (15q15.1-21.1, LGMD2A), dysferlin (2p13.3-13.1, LGMD2B), gamma-sarcoglycan (13q12, LGMD2C), alpha-sarcoglycan, also known as adhalin (17q12-q21.3, LGMD2D), beta-sarcoglycan (4q12, LGMD2E), delta-sarcoglycan (5q33-q34, LGMD2F), telethonin (17q11-q12, LGMD2G), E3-ubiquitin ligase (9q31-q34.1, LGMD2H), fukutin-related protein (19q13.3, LGMD2I), and titin (2q31, LGMD2J). Cardiac involvement has been described for LGMD1B-E, LGMD2C-G, and LGMD2I. The time of onset varies between early childhood and middle age. There is no male or female preponderance. Disease progression and life expectancy vary widely, even among different members of the same family. The diagnosis is based primarily on DNA analysis. The history, clinical neurological examinations, blood chemistry investigations, electromyography, and muscle biopsy also provide information that is helpful for the diagnosis. No causal therapy is currently available.
Collapse
Affiliation(s)
- J Finsterer
- Neurologische Abteilung, KA Rudolfstiftung, Wien, Osterreich.
| |
Collapse
|