1
|
Quinn CJ, Cartwright EJ, Trafford AW, Dibb KM. On the role of dysferlin in striated muscle: membrane repair, t-tubules and Ca 2+ handling. J Physiol 2024; 602:1893-1910. [PMID: 38615232 DOI: 10.1113/jp285103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 03/05/2024] [Indexed: 04/15/2024] Open
Abstract
Dysferlin is a 237 kDa membrane-associated protein characterised by multiple C2 domains with a diverse role in skeletal and cardiac muscle physiology. Mutations in DYSF are known to cause various types of human muscular dystrophies, known collectively as dysferlinopathies, with some patients developing cardiomyopathy. A myriad of in vitro membrane repair studies suggest that dysferlin plays an integral role in the membrane repair complex in skeletal muscle. In comparison, less is known about dysferlin in the heart, but mounting evidence suggests that dysferlin's role is similar in both muscle types. Recent findings have shown that dysferlin regulates Ca2+ handling in striated muscle via multiple mechanisms and that this becomes more important in conditions of stress. Maintenance of the transverse (t)-tubule network and the tight coordination of excitation-contraction coupling are essential for muscle contractility. Dysferlin regulates the maintenance and repair of t-tubules, and it is suspected that dysferlin regulates t-tubules and sarcolemmal repair through a similar mechanism. This review focuses on the emerging complexity of dysferlin's activity in striated muscle. Such insights will progress our understanding of the proteins and pathways that regulate basic heart and skeletal muscle function and help guide research into striated muscle pathology, especially that which arises due to dysferlin dysfunction.
Collapse
Affiliation(s)
- C J Quinn
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - E J Cartwright
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - A W Trafford
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| | - K M Dibb
- Unit of Cardiac Physiology, Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, 3.14 Core Technology Facility, Manchester, UK
| |
Collapse
|
2
|
Aguti S, Gallus GN, Bianchi S, Salvatore S, Rubegni A, Berti G, Formichi P, De Stefano N, Malandrini A, Lopergolo D. Novel Biomarkers for Limb Girdle Muscular Dystrophy (LGMD). Cells 2024; 13:329. [PMID: 38391941 PMCID: PMC10886967 DOI: 10.3390/cells13040329] [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: 12/15/2023] [Revised: 01/22/2024] [Accepted: 02/02/2024] [Indexed: 02/24/2024] Open
Abstract
OBJECTIVE To identify novel biomarkers as an alternative diagnostic tool for limb girdle muscular dystrophy (LGMD). BACKGROUND LGMD encompasses a group of muscular dystrophies characterized by proximal muscles weakness, elevated CK levels and dystrophic findings on muscle biopsy. Heterozygous CAPN3 mutations are associated with autosomal dominant LGMD-4, while biallelic mutations can cause autosomal recessive LGMD-1. Diagnosis is currently often based on invasive methods requiring muscle biopsy or blood tests. In most cases Western blotting (WB) analysis from muscle biopsy is essential for a diagnosis, as muscle samples are currently the only known tissues to express the full-length CAPN3 isoform. METHODS We analyzed CAPN3 in a cohort including 60 LGMD patients. Selected patients underwent a complete neurological examination, electromyography, muscle biopsy, and skin biopsies for primary fibroblasts isolation. The amount of CAPN3 was evaluated by WB analysis in muscle and skin tissues. The total RNA isolated from muscle, fibroblast and urine was processed, and cDNA was used for qualitative analysis. The expression of CAPN3 was investigated by qRT-PCR. The CAPN3 3D structure has been visualized and analyzed using PyMOL. RESULTS Among our patients, seven different CAPN3 mutations were detected, of which two were novel. After sequencing CAPN3 transcripts from fibroblast and urine, we detected different CAPN3 isoforms surprisingly including the full-length transcript. We found comparable protein levels from fibroblasts and muscle tissue; in particular, patients harboring a novel CAPN3 mutation showed a 30% reduction in protein compared to controls from both tissues. CONCLUSIONS Our findings showed for the first time the presence of the CAPN3 full-length transcript in urine and skin samples. Moreover, we demonstrated surprisingly comparable CAPN3 protein levels between muscle and skin samples, thus allowing us to hypothesize the use of skin biopsy and probably of urine samples as an alternative less invasive method to assess the amount of CAPN3 when molecular diagnosis turns out to be inconclusive.
Collapse
Affiliation(s)
- Sara Aguti
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (S.A.); (G.N.G.); (S.B.); (S.S.); (G.B.); (P.F.); (N.D.S.); (A.M.)
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Policlinico Le Scotte, Viale Bracci, 16, 53100 Siena, Italy
| | - Gian Nicola Gallus
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (S.A.); (G.N.G.); (S.B.); (S.S.); (G.B.); (P.F.); (N.D.S.); (A.M.)
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Policlinico Le Scotte, Viale Bracci, 16, 53100 Siena, Italy
| | - Silvia Bianchi
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (S.A.); (G.N.G.); (S.B.); (S.S.); (G.B.); (P.F.); (N.D.S.); (A.M.)
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Policlinico Le Scotte, Viale Bracci, 16, 53100 Siena, Italy
| | - Simona Salvatore
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (S.A.); (G.N.G.); (S.B.); (S.S.); (G.B.); (P.F.); (N.D.S.); (A.M.)
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Policlinico Le Scotte, Viale Bracci, 16, 53100 Siena, Italy
| | - Anna Rubegni
- Molecular Medicine for Neurodegenerative and Neuromuscular Disease Unit, IRCCS Stella Maris Foundation, 56128 Pisa, Italy;
| | - Gianna Berti
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (S.A.); (G.N.G.); (S.B.); (S.S.); (G.B.); (P.F.); (N.D.S.); (A.M.)
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Policlinico Le Scotte, Viale Bracci, 16, 53100 Siena, Italy
| | - Patrizia Formichi
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (S.A.); (G.N.G.); (S.B.); (S.S.); (G.B.); (P.F.); (N.D.S.); (A.M.)
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Policlinico Le Scotte, Viale Bracci, 16, 53100 Siena, Italy
| | - Nicola De Stefano
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (S.A.); (G.N.G.); (S.B.); (S.S.); (G.B.); (P.F.); (N.D.S.); (A.M.)
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Policlinico Le Scotte, Viale Bracci, 16, 53100 Siena, Italy
| | - Alessandro Malandrini
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (S.A.); (G.N.G.); (S.B.); (S.S.); (G.B.); (P.F.); (N.D.S.); (A.M.)
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Policlinico Le Scotte, Viale Bracci, 16, 53100 Siena, Italy
| | - Diego Lopergolo
- Department of Medicine, Surgery and Neurosciences, University of Siena, 53100 Siena, Italy; (S.A.); (G.N.G.); (S.B.); (S.S.); (G.B.); (P.F.); (N.D.S.); (A.M.)
- UOC Neurologia e Malattie Neurometaboliche, Azienda Ospedaliero-Universitaria Senese, Policlinico Le Scotte, Viale Bracci, 16, 53100 Siena, Italy
| |
Collapse
|
3
|
Mavrommatis L, Zaben A, Kindler U, Kienitz MC, Dietz J, Jeong HW, Böhme P, Brand-Saberi B, Vorgerd M, Zaehres H. CRISPR/Cas9 Genome Editing in LGMD2A/R1 Patient-Derived Induced Pluripotent Stem and Skeletal Muscle Progenitor Cells. Stem Cells Int 2023; 2023:9246825. [PMID: 38020204 PMCID: PMC10653971 DOI: 10.1155/2023/9246825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/16/2023] [Accepted: 06/08/2023] [Indexed: 12/01/2023] Open
Abstract
Large numbers of Calpain 3 (CAPN3) mutations cause recessive forms of limb-girdle muscular dystrophy (LGMD2A/LGMDR1) with selective atrophy of the proximal limb muscles. We have generated induced pluripotent stem cells (iPSC) from a patient with two mutations in exon 3 and exon 4 at the calpain 3 locus (W130C, 550delA). Two different strategies to rescue these mutations are devised: (i) on the level of LGMD2A-iPSC, we combined CRISPR/Cas9 genome targeting with a FACS and Tet transactivator-based biallelic selection strategy, which resulted in a new functional chimeric exon 3-4 without the two CAPN3 mutations. (ii) On the level of LGMD2A-iPSC-derived CD82+/Pax7+ myogenic progenitor cells, we demonstrate CRISPR/Cas9 mediated rescue of the highly prevalent exon 4 CAPN3 mutation. The first strategy specifically provides isogenic LGMD2A corrected iPSC for disease modelling, and the second strategy can be further elaborated for potential translational approaches.
Collapse
Affiliation(s)
- Lampros Mavrommatis
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular Embryology, 44801 Bochum, Germany
- Ruhr University Bochum, Medical Faculty, Department of Neurology with Heimer Institute for Muscle Research, University Hospital Bergmannsheil, 44789 Bochum, Germany
- Max Planck Institute for Molecular Biomedicine, Department of Cell and Developmental Biology, 48149 Münster, Germany
| | - Abdul Zaben
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular Embryology, 44801 Bochum, Germany
- Ruhr University Bochum, Medical Faculty, Department of Neurology with Heimer Institute for Muscle Research, University Hospital Bergmannsheil, 44789 Bochum, Germany
| | - Urs Kindler
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular Embryology, 44801 Bochum, Germany
| | - Marie-Cécile Kienitz
- Ruhr University Bochum, Medical Faculty, Department of Cellular Physiology, 44801 Bochum, Germany
| | - Julienne Dietz
- Ruhr University Bochum, Medical Faculty, Department of Neurology with Heimer Institute for Muscle Research, University Hospital Bergmannsheil, 44789 Bochum, Germany
- Witten/Herdecke University, Institute of Virology and Microbiology, Department of Human Medicine, Faculty of Health, 58453 Witten, Germany
| | - Hyun-Woo Jeong
- Max Planck Institute for Molecular Biomedicine, Sequencing Core Facility, 48149 Münster, Germany
| | - Pierre Böhme
- Ruhr University Bochum, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital Bochum, 44791 Bochum, Germany
| | - Beate Brand-Saberi
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular Embryology, 44801 Bochum, Germany
| | - Matthias Vorgerd
- Ruhr University Bochum, Medical Faculty, Department of Neurology with Heimer Institute for Muscle Research, University Hospital Bergmannsheil, 44789 Bochum, Germany
| | - Holm Zaehres
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular Embryology, 44801 Bochum, Germany
- Max Planck Institute for Molecular Biomedicine, Department of Cell and Developmental Biology, 48149 Münster, Germany
| |
Collapse
|
4
|
Rico A, Valls A, Guembelzu G, Azpitarte M, Aiastui A, Zufiria M, Jaka O, López de Munain A, Sáenz A. Altered expression of proteins involved in metabolism in LGMDR1 muscle is lost in cell culture conditions. Orphanet J Rare Dis 2023; 18:315. [PMID: 37817200 PMCID: PMC10565977 DOI: 10.1186/s13023-023-02873-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 08/24/2023] [Indexed: 10/12/2023] Open
Abstract
BACKGROUND Limb-girdle muscular dystrophy R1 calpain 3-related (LGMDR1) is an autosomal recessive muscular dystrophy due to mutations in the CAPN3 gene. While the pathophysiology of this disease has not been clearly established yet, Wnt and mTOR signaling pathways impairment in LGMDR1 muscles has been reported. RESULTS A reduction in Akt phosphorylation ratio and upregulated expression of proteins implicated in glycolysis (HK-II) and in fructose and lactate transport (GLUT5 and MCT1) in LGMDR1 muscle was observed. In vitro analysis to establish mitochondrial and glycolytic functions of primary cultures were performed, however, no differences between control and patients were observed. Additionally, gene expression analysis showed a lack of correlation between primary myoblasts/myotubes and LGMDR1 muscle while skin fibroblasts and CD56- cells showed a slightly better correlation with muscle. FRZB gene was upregulated in all the analyzed cell types (except in myoblasts). CONCLUSIONS Proteins implicated in metabolism are deregulated in LGMDR1 patients' muscle. Obtained results evidence the limited usefulness of primary myoblasts/myotubes for LGMDR1 gene expression and metabolic studies. However, since FRZB is the only gene that showed upregulation in all the analyzed cell types it is suggested its role as a key regulator of the pathophysiology of the LGMDR1 muscle fiber. The Wnt signaling pathway inactivation, secondary to FRZB upregulation, and GLUT5 overexpression may participate in the impaired adipogenesis in LGMD1R patients.
Collapse
Affiliation(s)
- Anabel Rico
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
| | - Andrea Valls
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
| | - Garazi Guembelzu
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
| | - Margarita Azpitarte
- Cell Culture, Histology and Multidisciplinary 3D Printing Platform, Biodonostia Health Research Institute, San Sebastián, Spain
| | - Ana Aiastui
- Department of Neurology, Donostialdea Integrated Health Organization, San Sebastián, Spain
| | - Mónica Zufiria
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
| | - Oihane Jaka
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
| | - Adolfo López de Munain
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain
- Department of Neurology, Donostialdea Integrated Health Organization, San Sebastián, Spain
- Department of Neurosciences, University of the Basque Country UPV-EHU, San Sebastián, Spain
- Faculty of Medicine, University of Deusto, Bilbao, Spain
| | - Amets Sáenz
- Neurosciences Area, Biodonostia Health Research Institute, San Sebastián, Spain.
- CIBERNED, CIBER, Spanish Ministry of Science and Innovation, Carlos III Health Institute, Madrid, Spain.
| |
Collapse
|
5
|
Arab F, Ahangari N, Malek H, Doosti M, Najarzadeh Torbati P, Ghayoor Karimiani E. Limb-Girdle Muscular Dystrophy Type 2B (LGMD2B) caused by Pathogenic Splice and Missense Variants of DYSF Gene among Iranians with Muscular Dystrophy. Adv Biomed Res 2023; 12:150. [PMID: 37564451 PMCID: PMC10410417 DOI: 10.4103/abr.abr_131_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 05/15/2022] [Accepted: 05/16/2022] [Indexed: 08/12/2023] Open
Abstract
Background The phenotypic range of limb-girdle muscular dystrophies (LGMDs) varies significantly because of genetic heterogeneity ranging from very mild to severe forms. Molecular analysis of the DYSF gene is challenging due to the wide range of mutations and associated complications in interpretations of novel DYSF variants with uncertain significance. Thus, in the current study, we performed the NGS analysis and its results are confirmed with Sanger sequencing to find the plausible disease-causing variants in patients with muscular dystrophy and their relatives via segregation analysis. Materials and Methods Nine patients with LGMD type 2B (LGMD2B) characteristics were screened for putative mutations by the whole-exome sequencing (WES) test. Either the patients themselves or their parents and first relatives were investigated in the segregation analysis through Sanger sequencing. The majority of variants were classified as pathogenic through American College of Medical Genetics and Genomics (ACMG) guidelines, segregation results, and in silico predictions. Results Results revealed eight variants in DYSF gene, including three splicing (c.1149+4A>G, c.2864+1G>A, and c.5785-7G>A), two nonsense (p.Gln112Ter and p.Trp2084Ter), two missense (p.Thr1546Pro and p.Tyr1032Cys), and one frameshift (p.Asp1067Ilefs), among nine Iranian families. One of the eight identified variants was novel, including p.Asp1067Ilefs, which was predicted to be likely pathogenic based on the ACMG guidelines. Notably, prediction tools suggested the damaging effects of studied variants on dysferlin structure. Conclusion Conclusively, the current report introduced eight variants including a novel frameshift in DYSF gene with noticeable pathogenic effects. This study significantly can broaden the diagnostic spectrum of LGMD2B in combination with previous reports about DYSF mutations and may pave the way for a rapidly high-ranked identification of the accurate type of dysferlinopathy.
Collapse
Affiliation(s)
- Fatemeh Arab
- Department of Medical Genetics, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Najmeh Ahangari
- Innovative Medical Research Center, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Hadis Malek
- Department of Medical Genetics, Next Generation Genetic Polyclinic, Mashhad, Iran
| | - Mohammad Doosti
- Department of Medical Genetics, Next Generation Genetic Polyclinic, Mashhad, Iran
| | | | - Ehsan Ghayoor Karimiani
- Department of Medical Genetics, Next Generation Genetic Polyclinic, Mashhad, Iran
- Molecular and Clinical Sciences Institute, St. George's University of London, Cranmer Terrace, London, United Kingdom, Iran
| |
Collapse
|
6
|
Allosteric Modulation of GSK-3β as a New Therapeutic Approach in Limb Girdle Muscular Dystrophy R1 Calpain 3-Related. Int J Mol Sci 2021; 22:ijms22147367. [PMID: 34298987 PMCID: PMC8308041 DOI: 10.3390/ijms22147367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/28/2021] [Accepted: 07/06/2021] [Indexed: 12/28/2022] Open
Abstract
Limb-girdle muscular dystrophy R1 calpain 3-related (LGMDR1) is an autosomal recessive muscular dystrophy produced by mutations in the CAPN3 gene. It is a rare disease and there is no cure or treatment for the disease while the pathophysiological mechanism by which the absence of calpain 3 provokes the dystrophy in muscles is not clear. However, key proteins implicated in Wnt and mTOR signaling pathways, which regulate muscle homeostasis, showed a considerable reduction in their expression and in their phosphorylation in LGMDR1 patients' muscles. Finally, the administration of tideglusib and VP0.7, ATP non-competitive inhibitors of glycogen synthase kinase 3β (GSK-3β), restore the expression and phosphorylation of these proteins in LGMDR1 cells, opening the possibility of their use as therapeutic options.
Collapse
|
7
|
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
|
8
|
Rekik S, Sakka S, Ben Romdhan S, Farhat N, Baba Amer Y, Lehkim L, Authier FJ, Mhiri C. Novel Missense CAPN3 Mutation Responsible for Adult-Onset Limb Girdle Muscular Dystrophy with Calves Hypertrophy. J Mol Neurosci 2019; 69:563-569. [PMID: 31410652 DOI: 10.1007/s12031-019-01383-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 07/09/2019] [Indexed: 02/01/2023]
Abstract
CAPN3 gene encodes for calpain-3; this protein is a calcium-dependent intracellular protease. Deficiency of this enzyme leads to weakness of the proximal limb muscles and pelvic and shoulder girdles, the so-called limb-girdle muscular dystrophy type 2A (LGMD2A). Here, we reported the case of a Tunisian patient with LGMD2A associated with a novel missense mutation (c.T1681C/p.Y561H). A 61-year-old man, with consanguineous parents, was referred for gait difficulties and slowly progressive proximal weakness of the four limbs associated with moderate hypertrophy of the calves but his facial muscles were unaffected. Electromyography showed that the profile was myopathic pattern and creatine kinase (CK) level was high. Muscle biopsy processing included routine histological, immunohistochemical, and Western Blot reactions, using a panel of antibodies directed against dystrophin, dysferlin, calpain-3, sarcoglycan α, β, γ, and δ. For mutation analysis, we designed an NGS-based screening. Immunological analyses demonstrated a total deficiency in calpain-3 and δ-sarcoglycan, and a reduced expression of dysferlin. The genetic study yielded a homozygous missense mutation (c.T1681C) of the 13th exon of the CAPN3 gene. The mutation found in our patient (c.T1681C/p.Y561H) has not been previously reported. It is responsible for complete calpain-3 and δ-sarcoglycan deficiency and reduced dysferlin expression. The genetic study is mandatory in such cases with multiple-protein deficiency and ambiguous results of immune-histology and Western Blot studies.
Collapse
Affiliation(s)
- Sabrine Rekik
- Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease (LR-12-SP-19), University Hospital Habib Bourguiba, Sfax, Tunisia. .,Clinical Investigation Center (CIC), CHU Habib Bourguiba, Sfax, Tunisia.
| | - Salma Sakka
- Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease (LR-12-SP-19), University Hospital Habib Bourguiba, Sfax, Tunisia
| | - Sawssan Ben Romdhan
- Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease (LR-12-SP-19), University Hospital Habib Bourguiba, Sfax, Tunisia.,Clinical Investigation Center (CIC), CHU Habib Bourguiba, Sfax, Tunisia
| | - Nouha Farhat
- Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease (LR-12-SP-19), University Hospital Habib Bourguiba, Sfax, Tunisia
| | - Yasmine Baba Amer
- U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, Créteil, France
| | - Leila Lehkim
- Anatomopathology Laboratory, CHU Habib Bourguiba, Sfax, Tunisia
| | | | - Chokri Mhiri
- Laboratory of Neurogenetics, Parkinson's Disease and Cerebrovascular Disease (LR-12-SP-19), University Hospital Habib Bourguiba, Sfax, Tunisia.,Clinical Investigation Center (CIC), CHU Habib Bourguiba, Sfax, Tunisia
| |
Collapse
|
9
|
Ishiba R, Santos ALF, Almeida CF, Caires LC, Ribeiro AF, Ayub-Guerrieri D, Fernandes SA, Souza LS, Vainzof M. Faster regeneration associated to high expression of Fam65b and Hdac6 in dysferlin-deficient mouse. J Mol Histol 2019; 50:375-387. [DOI: 10.1007/s10735-019-09834-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 06/10/2019] [Indexed: 11/27/2022]
|
10
|
|
11
|
Mapping QTL for white striping in relation to breast muscle yield and meat quality traits in broiler chickens. BMC Genomics 2018; 19:202. [PMID: 29554873 PMCID: PMC5859760 DOI: 10.1186/s12864-018-4598-9] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Accepted: 03/13/2018] [Indexed: 11/23/2022] Open
Abstract
Background White striping (WS) is an emerging muscular defect occurring on breast and thigh muscles of broiler chickens. It is characterized by the presence of white striations parallel to the muscle fibers and has significant consequences for meat quality. The etiology of WS remains poorly understood, even if previous studies demonstrated that the defect prevalence is related to broiler growth and muscle development. Moreover, recent studies showed moderate to high heritability values of WS, which emphasized the role of genetics in the expression of the muscle defect. The aim of this study was to identify the first quantitative trait loci (QTLs) for WS as well as breast muscle yield (BMY) and meat quality traits using a genome-wide association study (GWAS). We took advantage of two divergent lines of chickens selected for meat quality through Pectoralis major ultimate pH (pHu) and which exhibit the muscular defect. An expression QTL (eQTL) detection was further performed for some candidate genes, either suggested by GWAS analysis or based on their biological function. Results Forty-two single nucleotide polymorphisms (SNPs) associated with WS and other meat quality traits were identified. They defined 18 QTL regions located on 13 chromosomes. These results supported a polygenic inheritance of the studied traits and highlighted a few pleiotropic regions. A set of 16 positional and/or functional candidate genes was designed for further eQTL detection. A total of 132 SNPs were associated with molecular phenotypes and defined 21 eQTL regions located on 16 chromosomes. Interestingly, several co-localizations between QTL and eQTL regions were observed which could suggest causative genes and gene networks involved in the variability of meat quality traits and BMY. Conclusions The QTL mapping carried out in the current study for WS did not support the existence of a major gene, but rather suggested a polygenic inheritance of the defect and of other studied meat quality traits. We identified several candidate genes involved in muscle metabolism and structure and in muscular dystrophies. The eQTL analyses showed that they were part of molecular networks associated with WS and meat quality phenotypes and suggested a few putative causative genes. Electronic supplementary material The online version of this article (10.1186/s12864-018-4598-9) contains supplementary material, which is available to authorized users.
Collapse
|
12
|
An eccentric calpain, CAPN3/p94/calpain-3. Biochimie 2016; 122:169-87. [DOI: 10.1016/j.biochi.2015.09.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 09/07/2015] [Indexed: 01/09/2023]
|
13
|
Izumi R, Niihori T, Takahashi T, Suzuki N, Tateyama M, Watanabe C, Sugie K, Nakanishi H, Sobue G, Kato M, Warita H, Aoki Y, Aoki M. Genetic profile for suspected dysferlinopathy identified by targeted next-generation sequencing. NEUROLOGY-GENETICS 2015; 1:e36. [PMID: 27066573 PMCID: PMC4811388 DOI: 10.1212/nxg.0000000000000036] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 10/26/2015] [Indexed: 11/29/2022]
Abstract
Objective: To investigate the genetic causes of suspected dysferlinopathy and to reveal the genetic profile for myopathies with dysferlin deficiency. Methods: Using next-generation sequencing, we analyzed 42 myopathy-associated genes, including DYSF, in 64 patients who were clinically or pathologically suspected of having dysferlinopathy. Putative pathogenic mutations were confirmed by Sanger sequencing. In addition, copy-number variations in DYSF were investigated using multiplex ligation-dependent probe amplification. We also analyzed the genetic profile for 90 patients with myopathy with dysferlin deficiency, as indicated by muscle specimen immunohistochemistry, including patients from a previous cohort. Results: We identified putative pathogenic mutations in 38 patients (59% of all investigated patients). Twenty-three patients had DYSF mutations, including 6 novel mutations. The remaining 16 patients, including a single patient who also carried the DYSF mutation, harbored putative pathogenic mutations in other genes. The genetic profile for 90 patients with dysferlin deficiency revealed that 70% had DYSF mutations (n = 63), 10% had CAPN3 mutations (n = 9), 2% had CAV3 mutations (n = 2), 3% had mutations in other genes (in single patients), and 16% did not have any identified mutations (n = 14). Conclusions: This study clarified the heterogeneous genetic profile for myopathies with dysferlin deficiency. Our results demonstrate the importance of a comprehensive analysis of related genes in improving the genetic diagnosis of dysferlinopathy as one of the most common subtypes of limb-girdle muscular dystrophy. Unresolved diagnoses should be investigated using whole-genome or whole-exome sequencing.
Collapse
Affiliation(s)
- Rumiko Izumi
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tetsuya Niihori
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Toshiaki Takahashi
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Naoki Suzuki
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Maki Tateyama
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Chigusa Watanabe
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuma Sugie
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hirotaka Nakanishi
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Gen Sobue
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masaaki Kato
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hitoshi Warita
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yoko Aoki
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masashi Aoki
- Departments of Neurology (R.I., N.S., M.T., M.K., H.W., M.A.) and Medical Genetics (R.I., T.N., Y.A.), Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Neurology (T.T.), National Hospital Organization Sendai-Nishitaga, National Hospital, Sendai, Japan; Department of Neurology (M.T.), Iwate National Hospital, Ichinoseki, Japan; Department of Neurology (C.W.), Hiroshima-Nishi Medical Center, Hiroshima, Japan; Department of Neurology (K.S.), Nara Medical University, Nara, Japan; and Department of Neurology (H.N.) and Research Division for Neurodegeneration and Dementia (G.S.), Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
14
|
Dominov JA, Uyan O, Sapp PC, McKenna-Yasek D, Nallamilli BRR, Hegde M, Brown RH. A novel dysferlin mutant pseudoexon bypassed with antisense oligonucleotides. Ann Clin Transl Neurol 2014; 1:703-20. [PMID: 25493284 PMCID: PMC4241797 DOI: 10.1002/acn3.96] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Revised: 08/01/2014] [Accepted: 08/04/2014] [Indexed: 12/12/2022] Open
Abstract
Objective Mutations in dysferlin (DYSF), a Ca2+-sensitive ferlin family protein important for membrane repair, vesicle trafficking, and T-tubule function, cause Miyoshi myopathy, limb-girdle muscular dystrophy type 2B, and distal myopathy. More than 330 pathogenic DYSF mutations have been identified within exons or near exon–intron junctions. In ~17% of patients who lack normal DYSF, only a single disease-causing mutation has been identified. We studied one family with one known mutant allele to identify both the second underlying genetic defect and potential therapeutic approaches. Methods We sequenced the full DYSF cDNA and investigated antisense oligonucleotides (AONs) as a tool to modify splicing of the mRNA transcripts in order to process out mutant sequences. Results We identified a novel pseudoexon between exons 44 and 45, (pseudoexon 44.1, PE44.1), which inserts an additional 177 nucleotides into the mRNA and 59 amino acids within the conserved C2F domain of the DYSF protein. Two unrelated dysferlinopathy patients were also found to carry this mutation. Using AONs targeting PE44.1, we blocked the abnormal splicing event, yielding normal, full-length DYSF mRNA, and increased DYSF protein expression. Interpretation This is the first report of a deep intronic mutation in DYSF that alters mRNA splicing to include a mutant peptide fragment within a key DYSF domain. We report that AON-mediated exon-skipping restores production of normal, full-length DYSF in patients’ cells in vitro, offering hope that this approach will be therapeutic in this genetic context, and providing a foundation for AON therapeutics targeting other pathogenic DYSF alleles.
Collapse
Affiliation(s)
- Janice A Dominov
- Neurology Department, University of Massachusetts Medical School Worcester, Massachusetts, 01605
| | - Ozgün Uyan
- Neurology Department, University of Massachusetts Medical School Worcester, Massachusetts, 01605
| | - Peter C Sapp
- Neurology Department, University of Massachusetts Medical School Worcester, Massachusetts, 01605
| | - Diane McKenna-Yasek
- Neurology Department, University of Massachusetts Medical School Worcester, Massachusetts, 01605
| | - Babi R R Nallamilli
- Department of Human Genetics, Emory University School of Medicine Atlanta, Georgia, 30322
| | - Madhuri Hegde
- Department of Human Genetics, Emory University School of Medicine Atlanta, Georgia, 30322
| | - Robert H Brown
- Neurology Department, University of Massachusetts Medical School Worcester, Massachusetts, 01605
| |
Collapse
|
15
|
Jia X, Schulte L, Loukas A, Pickering D, Pearson M, Mobli M, Jones A, Rosengren KJ, Daly NL, Gobert GN, Jones MK, Craik DJ, Mulvenna J. Solution structure, membrane interactions, and protein binding partners of the tetraspanin Sm-TSP-2, a vaccine antigen from the human blood fluke Schistosoma mansoni. J Biol Chem 2014; 289:7151-7163. [PMID: 24429291 DOI: 10.1074/jbc.m113.531558] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The tetraspanins (TSPs) are a family of integral membrane proteins that are ubiquitously expressed at the surface of eukaryotic cells. TSPs mediate a range of processes at the surface of the plasma membrane by providing a scaffold for the assembly of protein complexes known as tetraspanin-enriched microdomains (TEMs). We report here the structure of the surface-exposed EC2 domain from Sm-TSP-2, a TSP from Schistosoma mansoni and one of the better prospects for the development of a vaccine against schistosomiasis. This is the first solution structure of this domain, and our investigations of its interactions with lipid micelles provide a general model for interactions between TSPs, membranes, and other proteins. Using chemical cross-linking, eight potential protein constituents of Sm-TSP-2-mediated TEMs were also identified. These include proteins important for membrane maintenance and repair, providing further evidence for the functional role of Sm-TSP-2- and Sm-TSP-2-mediated TEMs. The identification of calpain, Sm29, and fructose-bisphosphate aldolase, themselves potential vaccine antigens, suggests that the Sm-TSP-2-mediated TEMs could be disrupted via multiple targets. The identification of further Sm-TSP-2-mediated TEM proteins increases the available candidates for multiplex vaccines and/or novel drugs targeting TEMs in the schistosome tegument.
Collapse
Affiliation(s)
- Xinying Jia
- Queensland Institute of Medical Research, Brisbane, QLD 4006, Australia
| | - Leigh Schulte
- Queensland Institute of Medical Research, Brisbane, QLD 4006, Australia; The University of Queensland, School of Veterinary Sciences, Gatton, QLD 4343, Australia
| | - Alex Loukas
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, James Cook University, Cairns, QLD 4878, Australia
| | - Darren Pickering
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, James Cook University, Cairns, QLD 4878, Australia
| | - Mark Pearson
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, James Cook University, Cairns, QLD 4878, Australia
| | - Mehdi Mobli
- The University of Queensland, Centre for Advanced Imaging, Brisbane, QLD 4072, Australia
| | - Alun Jones
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, QLD 4072, Australia
| | - Karl J Rosengren
- The University of Queensland, School of Biomedical Sciences, Brisbane, QLD 4072, Australia
| | - Norelle L Daly
- Centre for Biodiscovery and Molecular Development of Therapeutics, Queensland Tropical Health Alliance, James Cook University, Cairns, QLD 4878, Australia
| | - Geoffrey N Gobert
- Queensland Institute of Medical Research, Brisbane, QLD 4006, Australia
| | - Malcolm K Jones
- Queensland Institute of Medical Research, Brisbane, QLD 4006, Australia; The University of Queensland, School of Veterinary Sciences, Gatton, QLD 4343, Australia
| | - David J Craik
- The University of Queensland, Institute for Molecular Bioscience, Brisbane, QLD 4072, Australia
| | - Jason Mulvenna
- Queensland Institute of Medical Research, Brisbane, QLD 4006, Australia; The University of Queensland, School of Biomedical Sciences, Brisbane, QLD 4072, Australia.
| |
Collapse
|
16
|
Pyatnitskiy M, Mazo I, Shkrob M, Schwartz E, Kotelnikova E. Clustering gene expression regulators: new approach to disease subtyping. PLoS One 2014; 9:e84955. [PMID: 24416320 PMCID: PMC3887006 DOI: 10.1371/journal.pone.0084955] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2013] [Accepted: 11/20/2013] [Indexed: 12/29/2022] Open
Abstract
One of the main challenges in modern medicine is to stratify different patient groups in terms of underlying disease molecular mechanisms as to develop more personalized approach to therapy. Here we propose novel method for disease subtyping based on analysis of activated expression regulators on a sample-by-sample basis. Our approach relies on Sub-Network Enrichment Analysis algorithm (SNEA) which identifies gene subnetworks with significant concordant changes in expression between two conditions. Subnetwork consists of central regulator and downstream genes connected by relations extracted from global literature-extracted regulation database. Regulators found in each patient separately are clustered together and assigned activity scores which are used for final patients grouping. We show that our approach performs well compared to other related methods and at the same time provides researchers with complementary level of understanding of pathway-level biology behind a disease by identification of significant expression regulators. We have observed the reasonable grouping of neuromuscular disorders (triggered by structural damage vs triggered by unknown mechanisms), that was not revealed using standard expression profile clustering. For another experiment we were able to suggest the clusters of regulators, responsible for colorectal carcinoma vs adenoma discrimination and identify frequently genetically changed regulators that could be of specific importance for the individual characteristics of cancer development. Proposed approach can be regarded as biologically meaningful feature selection, reducing tens of thousands of genes down to dozens of clusters of regulators. Obtained clusters of regulators make possible to generate valuable biological hypotheses about molecular mechanisms related to a clinical outcome for individual patient.
Collapse
Affiliation(s)
- Mikhail Pyatnitskiy
- Institute of Biomedical Chemistry, RAMS, Moscow, Russia
- Ariadne Diagnostics LLC, Rockville, Maryland, United States of America
- * E-mail:
| | - Ilya Mazo
- Ariadne Diagnostics LLC, Rockville, Maryland, United States of America
| | - Maria Shkrob
- Elsevier Inc, Rockville, Maryland, United States of America
| | - Elena Schwartz
- Ariadne Diagnostics LLC, Rockville, Maryland, United States of America
| | - Ekaterina Kotelnikova
- Ariadne Diagnostics LLC, Rockville, Maryland, United States of America
- Institute for Information Transmission Problems, RAS, Moscow, Russia
| |
Collapse
|
17
|
Flix B, de la Torre C, Castillo J, Casal C, Illa I, Gallardo E. Dysferlin interacts with calsequestrin-1, myomesin-2 and dynein in human skeletal muscle. Int J Biochem Cell Biol 2013; 45:1927-38. [PMID: 23792176 DOI: 10.1016/j.biocel.2013.06.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Revised: 05/24/2013] [Accepted: 06/09/2013] [Indexed: 11/25/2022]
Abstract
Dysferlinopathies are a group of progressive muscular dystrophies characterized by mutations in the gene DYSF. These mutations cause scarcity or complete absence of dysferlin, a protein that is expressed in skeletal muscle and plays a role in membrane repair. Our objective was to unravel the proteins that constitute the dysferlin complex and their interaction within the complex using immunoprecipitation assays (IP), blue native gel electrophoresis (BN) in healthy adult skeletal muscle and healthy cultured myotubes, and fluorescence lifetime imaging-fluorescence resonance energy transfer (FLIM-FRET) analysis in healthy myotubes. The combination of immunoprecipitations and blue native electrophoresis allowed us to identify previously reported partners of dysferlin - such as caveolin-3, AHNAK, annexins, or Trim72/MG53 - and new interacting partners. Fluorescence lifetime imaging showed a direct interaction of dysferlin with Trim72/MG53, AHNAK, cytoplasmic dynein, myomesin-2 and calsequestrin-1, but not with caveolin-3 or dystrophin. In conclusion, although IP and BN are useful tools to identify the proteins in a complex, techniques such as fluorescence lifetime imaging analysis are needed to determine the direct and indirect interactions of these proteins within the complex. This knowledge may help us to better understand the roles of dysferlin in muscle tissue and identify new genes involved in muscular dystrophies in which the responsible gene is unknown.
Collapse
Affiliation(s)
- Bàrbara Flix
- Servei de Neurologia, Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Barcelona, Spain
| | | | | | | | | | | |
Collapse
|
18
|
de Morrée A, Flix B, Bagaric I, Wang J, van den Boogaard M, Grand Moursel L, Frants RR, Illa I, Gallardo E, Toes R, van der Maarel SM. Dysferlin regulates cell adhesion in human monocytes. J Biol Chem 2013; 288:14147-14157. [PMID: 23558685 DOI: 10.1074/jbc.m112.448589] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Dysferlin is mutated in a group of muscular dystrophies commonly referred to as dysferlinopathies. It is highly expressed in skeletal muscle, where it is important for sarcolemmal maintenance. Recent studies show that dysferlin is also expressed in monocytes. Moreover, muscle of dysferlinopathy patients is characterized by massive immune cell infiltrates, and dysferlin-negative monocytes were shown to be more aggressive and phagocytose more particles. This suggests that dysferlin deregulation in monocytes might contribute to disease progression, but the molecular mechanism is unclear. Here we show that dysferlin expression is increased with differentiation in human monocytes and the THP1 monocyte cell model. Freshly isolated monocytes of dysferlinopathy patients show deregulated expression of fibronectin and fibronectin-binding integrins, which is recapitulated by transient knockdown of dysferlin in THP1 cells. Dysferlin forms a protein complex with these integrins at the cell membrane, and its depletion impairs cell adhesion. Moreover, patient macrophages show altered adhesion and motility. These findings suggest that dysferlin is involved in regulating cellular interactions and provide new insight into dysferlin function in inflammatory cells.
Collapse
Affiliation(s)
- Antoine de Morrée
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Bàrbara Flix
- Servei de Neurologia, Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Universitat Autònoma de Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08193 Bellaterra, Spain
| | - Ivana Bagaric
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Jun Wang
- Department of Rheumatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | | | - Laure Grand Moursel
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Rune R Frants
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Isabel Illa
- Servei de Neurologia, Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Universitat Autònoma de Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08193 Bellaterra, Spain
| | - Eduard Gallardo
- Servei de Neurologia, Laboratori de Neurologia Experimental, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Universitat Autònoma de Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), 08193 Bellaterra, Spain
| | - Rene Toes
- Department of Rheumatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Silvère M van der Maarel
- Department of Human Genetics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands.
| |
Collapse
|
19
|
Sarkozy A, Bushby K, Mercuri E. Muscular Dystrophies. EMERY AND RIMOIN'S PRINCIPLES AND PRACTICE OF MEDICAL GENETICS 2013:1-58. [DOI: 10.1016/b978-0-12-383834-6.00134-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
20
|
Tabebordbar M, Wang ET, Wagers AJ. Skeletal muscle degenerative diseases and strategies for therapeutic muscle repair. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2012; 8:441-75. [PMID: 23121053 DOI: 10.1146/annurev-pathol-011811-132450] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Skeletal muscle is a highly specialized, postmitotic tissue that must withstand chronic mechanical and physiological stress throughout life to maintain proper contractile function. Muscle damage or disease leads to progressive weakness and disability, and manifests in more than 100 different human disorders. Current therapies to treat muscle degenerative diseases are limited mostly to the amelioration of symptoms, although promising new therapeutic directions are emerging. In this review, we discuss the pathological basis for the most common muscle degenerative diseases and highlight new and encouraging experimental and clinical opportunities to prevent or reverse these afflictions.
Collapse
Affiliation(s)
- Mohammadsharif Tabebordbar
- Department of Stem Cell and Regenerative Biology, Harvard University and Harvard Stem Cell Institute, Cambridge, MA 02138, USA.
| | | | | |
Collapse
|
21
|
Lostal W, Bartoli M, Roudaut C, Bourg N, Krahn M, Pryadkina M, Borel P, Suel L, Roche JA, Stockholm D, Bloch RJ, Levy N, Bashir R, Richard I. Lack of correlation between outcomes of membrane repair assay and correction of dystrophic changes in experimental therapeutic strategy in dysferlinopathy. PLoS One 2012; 7:e38036. [PMID: 22666441 PMCID: PMC3362551 DOI: 10.1371/journal.pone.0038036] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 04/30/2012] [Indexed: 01/31/2023] Open
Abstract
Mutations in the dysferlin gene are the cause of Limb-girdle Muscular Dystrophy type 2B and Miyoshi Myopathy. The dysferlin protein has been implicated in sarcolemmal resealing, leading to the idea that the pathophysiology of dysferlin deficiencies is due to a deficit in membrane repair. Here, we show using two different approaches that fullfiling membrane repair as asseyed by laser wounding assay is not sufficient for alleviating the dysferlin deficient pathology. First, we generated a transgenic mouse overexpressing myoferlin to test the hypothesis that myoferlin, which is homologous to dysferlin, can compensate for the absence of dysferlin. The myoferlin overexpressors show no skeletal muscle abnormalities, and crossing them with a dysferlin-deficient model rescues the membrane fusion defect present in dysferlin-deficient mice in vitro. However, myoferlin overexpression does not correct muscle histology in vivo. Second, we report that AAV-mediated transfer of a minidysferlin, previously shown to correct the membrane repair deficit in vitro, also fails to improve muscle histology. Furthermore, neither myoferlin nor the minidysferlin prevented myofiber degeneration following eccentric exercise. Our data suggest that the pathogenicity of dysferlin deficiency is not solely related to impairment in sarcolemmal repair and highlight the care needed in selecting assays to assess potential therapies for dysferlinopathies.
Collapse
Affiliation(s)
| | - Marc Bartoli
- Département de Génétique Médicale, Hôpital d’Enfants de la Timone, AP-HM, and Inserm UMR_S 910, Faculté de Médecine Timone, Université de la Méditerranée, Marseille, France
| | | | | | - Martin Krahn
- Département de Génétique Médicale, Hôpital d’Enfants de la Timone, AP-HM, and Inserm UMR_S 910, Faculté de Médecine Timone, Université de la Méditerranée, Marseille, France
| | | | | | | | - Joseph A. Roche
- Department of Physiology, University of Maryland, School of Medicine, Baltimore, Maryland, United States of America
| | | | - Robert J. Bloch
- Department of Physiology, University of Maryland, School of Medicine, Baltimore, Maryland, United States of America
| | - Nicolas Levy
- Département de Génétique Médicale, Hôpital d’Enfants de la Timone, AP-HM, and Inserm UMR_S 910, Faculté de Médecine Timone, Université de la Méditerranée, Marseille, France
| | - Rumaisa Bashir
- School of Biological and Biomedical Sciences, University of Durham, Durham, United Kingdom
| | | |
Collapse
|
22
|
Gallardo E, de Luna N, Diaz-Manera J, Rojas-García R, Gonzalez-Quereda L, Flix B, de Morrée A, van der Maarel S, Illa I. Comparison of dysferlin expression in human skeletal muscle with that in monocytes for the diagnosis of dysferlin myopathy. PLoS One 2011; 6:e29061. [PMID: 22194990 PMCID: PMC3241698 DOI: 10.1371/journal.pone.0029061] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 11/20/2011] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Dysferlinopathies are caused by mutations in the dysferlin gene (DYSF). Diagnosis is complex due to the high clinical variability of the disease and because dysferlin expression in the muscle biopsy may be secondarily reduced due to a primary defect in some other gene. Dysferlin is also expressed in peripheral blood monocytes (PBM). Studying dysferlin in monocytes is used for the diagnosis of dysferlin myopathies. The aim of the study was to determine whether dysferlin expression in PBM correlates with that in skeletal muscle. METHODOLOGY/PRINCIPAL FINDINGS Using western-blot (WB) we quantified dysferlin expression in PBM from 21 pathological controls with other myopathies in whom mutations in DYSF were excluded and from 17 patients who had dysferlinopathy and two mutations in DYSF. Results were compared with protein expression in muscle by WB and immunohistochemistry (IH). We found a good correlation between skeletal muscle and monocytes using WB. However, IH results were misleading because abnormal expression of dysferlin was also observed in 13/21 pathological controls. CONCLUSIONS/SIGNIFICANCE The analysis of dysferlin protein expression in PBM is helpful when: 1) the skeletal muscle IH pattern is abnormal or 2) when muscle WB can not be performed either because muscle sample is lacking or insufficient or because the muscle biopsy is taken from a muscle at an end-stage and it mainly consists of fat and fibrotic tissue.
Collapse
Affiliation(s)
- Eduard Gallardo
- Servei de Neurologia, Laboratori de Malalties Neuromusculars, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Universitat Autònoma de Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Noemi de Luna
- Servei de Neurologia, Laboratori de Malalties Neuromusculars, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Universitat Autònoma de Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Jordi Diaz-Manera
- Servei de Neurologia, Laboratori de Malalties Neuromusculars, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Universitat Autònoma de Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Ricardo Rojas-García
- Servei de Neurologia, Laboratori de Malalties Neuromusculars, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Universitat Autònoma de Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Lidia Gonzalez-Quereda
- Servei de Genètica, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Universitat Autònoma and Centro de Investigación en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| | - Bàrbara Flix
- Servei de Neurologia, Laboratori de Malalties Neuromusculars, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Universitat Autònoma de Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
| | - Antoine de Morrée
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | | | - Isabel Illa
- Servei de Neurologia, Laboratori de Malalties Neuromusculars, Hospital de la Santa Creu i Sant Pau i Institut de Recerca de HSCSP, Universitat Autònoma de Barcelona and Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Barcelona, Spain
- * E-mail:
| |
Collapse
|
23
|
Humphrey GW, Mekhedov E, Blank PS, de Morree A, Pekkurnaz G, Nagaraju K, Zimmerberg J. GREG cells, a dysferlin-deficient myogenic mouse cell line. Exp Cell Res 2011; 318:127-35. [PMID: 22020321 DOI: 10.1016/j.yexcr.2011.10.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2011] [Revised: 10/03/2011] [Accepted: 10/04/2011] [Indexed: 11/29/2022]
Abstract
The dysferlinopathies (e.g. LGMD2b, Myoshi myopathy) are progressive, adult-onset muscle wasting syndromes caused by mutations in the gene coding for dysferlin. Dysferlin is a large (~200kDa) membrane-anchored protein, required for maintenance of plasmalemmal integrity in muscle fibers. To facilitate analysis of dysferlin function in muscle cells, we have established a dysferlin-deficient myogenic cell line (GREG cells) from the A/J mouse, a genetic model for dysferlinopathy. GREG cells have no detectable dysferlin expression, but proliferate normally in growth medium and fuse into functional myotubes in differentiation medium. GREG myotubes exhibit deficiencies in plasma membrane repair, as measured by laser wounding in the presence of FM1-43 dye. Under the wounding conditions used, the majority (~66%) of GREG myotubes lack membrane repair capacity, while no membrane repair deficiency was observed in dysferlin-normal C2C12 myotubes, assayed under the same conditions. We discuss the possibility that the observed heterogeneity in membrane resealing represents genetic compensation for dysferlin deficiency.
Collapse
Affiliation(s)
- Glen W Humphrey
- Program in Physical Biology, Eunice Kennedy Schriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Kawahara G, Serafini PR, Myers JA, Alexander MS, Kunkel LM. Characterization of zebrafish dysferlin by morpholino knockdown. Biochem Biophys Res Commun 2011; 413:358-63. [PMID: 21893049 DOI: 10.1016/j.bbrc.2011.08.105] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Accepted: 08/20/2011] [Indexed: 10/17/2022]
Abstract
Mutations in the gene encoding dysferlin cause two distinct muscular dystrophy phenotypes: limb-girdle muscular dystrophy type 2B (LGMD-2B) and Miyoshi myopathy (MM). Dysferlin is a large transmembrane protein involved in myoblast fusion and membrane resealing. Zebrafish represent an ideal animal model to use for studying muscle disease including abnormalities of dysferlin. cDNAs of zebrafish dysferlin were cloned (6.3 kb) and the predicted amino acid sequences, showed 68% similarity to predicted amino acid sequences of mammalian dysferlin. The expression of dysferlin was mainly in skeletal muscle, heart and eye, and the expression could be detected as early as 11h post fertilization (hpf). Three different antisense oligonucleotide morpholinos were targeted to inhibit translation of this dysferlin mRNA and the morpholino-injected fish showed marked muscle disorganization which could be detected by birefringence assay. Western blot analysis using dysferlin antibodies showed that the expression of dysferlin was reduced in each of the three morphants. Dysferlin expression was shown to be reduced at the myosepta of zebrafish muscle using immunohistochemistry, although the expression of other muscle membrane components, dystrophin, laminin, β-dystroglycan were detected normally. Our data suggest that zebrafish dysferlin expression is involved in stabilizing muscle structures and its downregulation causes muscle disorganization.
Collapse
Affiliation(s)
- Genri Kawahara
- Division of Genetics, Program in Genomics, Children's Hospital Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
25
|
172nd ENMC International Workshop: dysferlinopathies 29-31 January 2010, Naarden, The Netherlands. Neuromuscul Disord 2011; 21:503-12. [PMID: 21602046 DOI: 10.1016/j.nmd.2011.04.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 04/06/2011] [Accepted: 04/15/2011] [Indexed: 11/24/2022]
|
26
|
Martini P, Risso D, Sales G, Romualdi C, Lanfranchi G, Cagnin S. Statistical Test of Expression Pattern (STEPath): a new strategy to integrate gene expression data with genomic information in individual and meta-analysis studies. BMC Bioinformatics 2011; 12:92. [PMID: 21481242 PMCID: PMC3094239 DOI: 10.1186/1471-2105-12-92] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 04/11/2011] [Indexed: 01/21/2023] Open
Abstract
Background In the last decades, microarray technology has spread, leading to a dramatic increase of publicly available datasets. The first statistical tools developed were focused on the identification of significant differentially expressed genes. Later, researchers moved toward the systematic integration of gene expression profiles with additional biological information, such as chromosomal location, ontological annotations or sequence features. The analysis of gene expression linked to physical location of genes on chromosomes allows the identification of transcriptionally imbalanced regions, while, Gene Set Analysis focuses on the detection of coordinated changes in transcriptional levels among sets of biologically related genes. In this field, meta-analysis offers the possibility to compare different studies, addressing the same biological question to fully exploit public gene expression datasets. Results We describe STEPath, a method that starts from gene expression profiles and integrates the analysis of imbalanced region as an a priori step before performing gene set analysis. The application of STEPath in individual studies produced gene set scores weighted by chromosomal activation. As a final step, we propose a way to compare these scores across different studies (meta-analysis) on related biological issues. One complication with meta-analysis is batch effects, which occur because molecular measurements are affected by laboratory conditions, reagent lots and personnel differences. Major problems occur when batch effects are correlated with an outcome of interest and lead to incorrect conclusions. We evaluated the power of combining chromosome mapping and gene set enrichment analysis, performing the analysis on a dataset of leukaemia (example of individual study) and on a dataset of skeletal muscle diseases (meta-analysis approach). In leukaemia, we identified the Hox gene set, a gene set closely related to the pathology that other algorithms of gene set analysis do not identify, while the meta-analysis approach on muscular disease discriminates between related pathologies and correlates similar ones from different studies. Conclusions STEPath is a new method that integrates gene expression profiles, genomic co-expressed regions and the information about the biological function of genes. The usage of the STEPath-computed gene set scores overcomes batch effects in the meta-analysis approaches allowing the direct comparison of different pathologies and different studies on a gene set activation level.
Collapse
Affiliation(s)
- Paolo Martini
- CRIBI Biotechnology Centre, Department of Biology, University of Padova, via U, Bassi 58/B, 35121 Padova, Italy
| | | | | | | | | | | |
Collapse
|
27
|
|
28
|
Cacciottolo M, Belcastro V, Laval S, Bushby K, di Bernardo D, Nigro V. Reverse engineering gene network identifies new dysferlin-interacting proteins. J Biol Chem 2011; 286:5404-13. [PMID: 21119217 PMCID: PMC3037653 DOI: 10.1074/jbc.m110.173559] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2010] [Revised: 11/29/2010] [Indexed: 01/28/2023] Open
Abstract
Dysferlin (DYSF) is a type II transmembrane protein implicated in surface membrane repair of muscle. Mutations in dysferlin lead to Limb Girdle Muscular Dystrophy 2B (LGMD2B), Miyoshi Myopathy (MM), and Distal Myopathy with Anterior Tibialis onset (DMAT). The DYSF protein complex is not well understood, and only a few protein-binding partners have been identified thus far. To increase the set of interacting protein partners for DYSF we recovered a list of predicted interacting protein through a systems biology approach. The predictions are part of a "reverse-engineered" genome-wide human gene regulatory network obtained from experimental data by computational analysis. The reverse-engineering algorithm behind the analysis relates genes to each other based on changes in their expression patterns. DYSF and AHNAK were used to query the system and extract lists of potential interacting proteins. Among the 32 predictions the two genes share, we validated the physical interaction between DYSF protein with moesin (MSN) and polymerase I and transcript release factor (PTRF) in mouse heart lysate, thus identifying two novel Dysferlin-interacting proteins. Our strategy could be useful to clarify Dysferlin function in intracellular vesicles and its implication in muscle membrane resealing.
Collapse
Affiliation(s)
- Mafalda Cacciottolo
- From the TIGEM-Telethon Institute of Genetics and Medicine, 80131 Naples, Italy
| | - Vincenzo Belcastro
- From the TIGEM-Telethon Institute of Genetics and Medicine, 80131 Naples, Italy
| | - Steve Laval
- the Institute of Human Genetics, Newcastle University, NE1 3BZ Newcastle Upon Tyne, United Kingdom, and
| | - Kate Bushby
- the Institute of Human Genetics, Newcastle University, NE1 3BZ Newcastle Upon Tyne, United Kingdom, and
| | - Diego di Bernardo
- From the TIGEM-Telethon Institute of Genetics and Medicine, 80131 Naples, Italy
| | - Vincenzo Nigro
- From the TIGEM-Telethon Institute of Genetics and Medicine, 80131 Naples, Italy
- the Laboratorio di Genetica Medica, Dipartimento di Patologia Generale and CIRM, Seconda Università degli Studi di Napoli, 80138 Naples, Italy
| |
Collapse
|
29
|
van Haagen HHHBM, 't Hoen PAC, de Morrée A, van Roon-Mom WMC, Peters DJM, Roos M, Mons B, van Ommen GJ, Schuemie MJ. In silico discovery and experimental validation of new protein-protein interactions. Proteomics 2011; 11:843-53. [DOI: 10.1002/pmic.201000398] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Revised: 10/17/2010] [Accepted: 11/25/2010] [Indexed: 01/27/2023]
|
30
|
Abstract
Myoblast fusion contributes to muscle growth in development and during regeneration of mature muscle. Myoblasts fuse to each other as well as to multinucleate myotubes to enlarge the myofiber. The molecular mechanisms of myoblast fusion are incompletely understood. Adhesion, apposition, and membrane fusion are accompanied by cytoskeletal rearrangements. The ferlin family of proteins is implicated in human muscle disease and has been implicated in fusion events in muscle, including myoblast fusion, vesicle trafficking and membrane repair. Dysferlin was the first mammalian ferlin identified and it is now known that there are six different ferlins. Loss-of-function mutations in the dysferlin gene lead to limb girdle muscular dystrophy and the milder disorder Miyoshi Myopathy. Dysferlin is a membrane-associated protein that has been implicated in resealing disruptions in the muscle plasma membrane. Newer data supports a broader role for dysferlin in intracellular vesicular movement, a process also important for resealing. Myoferlin is highly expressed in myoblasts that undergoing fusion, and the absence of myoferlin leads to impaired myoblast fusion. Myoferlin also regulates intracellular trafficking events, including endocytic recycling, a process where internalized vesicles are returned to the plasma membrane. The trafficking role of ferlin proteins is reviewed herein with a specific focus as to how this machinery alters myogenesis and muscle growth.
Collapse
Affiliation(s)
- Avery D Posey
- Genomics and Systems Biology, Committee on Genetics, The University of Chicago, Chicago, Illinois, USA
| | | | | |
Collapse
|
31
|
Proteomic analysis of the dysferlin protein complex unveils its importance for sarcolemmal maintenance and integrity. PLoS One 2010; 5:e13854. [PMID: 21079765 PMCID: PMC2974636 DOI: 10.1371/journal.pone.0013854] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2010] [Accepted: 10/15/2010] [Indexed: 11/19/2022] Open
Abstract
Dysferlin is critical for repair of muscle membranes after damage. Mutations in dysferlin lead to a progressive muscular dystrophy. Recent studies suggest additional roles for dysferlin. We set out to study dysferlin's protein-protein interactions to obtain comprehensive knowledge of dysferlin functionalities in a myogenic context. We developed a robust and reproducible method to isolate dysferlin protein complexes from cells and tissue. We analyzed the composition of these complexes in cultured myoblasts, myotubes and skeletal muscle tissue by mass spectrometry and subsequently inferred potential protein functions through bioinformatics analyses. Our data confirm previously reported interactions and support a function for dysferlin as a vesicle trafficking protein. In addition novel potential functionalities were uncovered, including phagocytosis and focal adhesion. Our data reveal that the dysferlin protein complex has a dynamic composition as a function of myogenic differentiation. We provide additional experimental evidence and show dysferlin localization to, and interaction with the focal adhesion protein vinculin at the sarcolemma. Finally, our studies reveal evidence for cross-talk between dysferlin and its protein family member myoferlin. Together our analyses show that dysferlin is not only a membrane repair protein but also important for muscle membrane maintenance and integrity.
Collapse
|
32
|
Rosales XQ, Gastier-Foster JM, Lewis S, Vinod M, Thrush DL, Astbury C, Pyatt R, Reshmi S, Sahenk Z, Mendell JR. Novel diagnostic features of dysferlinopathies. Muscle Nerve 2010; 42:14-21. [PMID: 20544924 PMCID: PMC3025537 DOI: 10.1002/mus.21650] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Reports of dysferlinopathy have suggested a clinically heterogeneous group of patients. We identified specific novel molecular and phenotypic features that help distinguish dysferlinopathies from other forms of limb-girdle muscular dystrophy (LGMD). A detailed history, physical exam, and protein and mutation analysis of genomic DNA was done for all subjects. Five of 21 confirmed DYSF gene mutations were not previously reported. A distinct "bulge" of the deltoid muscle in combination with other findings was a striking feature in all patients. Six subjects had atypical calf enlargement, and 3 of these exhibited a paradoxical pattern of dysferlin expression: severely reduced by direct immunofluorescence with overexpression on Western blots. Six patients showed amyloid deposits in muscle that extended these findings to new domains of the dysferlin gene, including the C2G domain. Correlative studies showed colocalization of amyloid with deposition of dysferlin. The present data further serve to guide clinicians facing the expensive task of molecular characterization of patients with an LGMD phenotype.
Collapse
Affiliation(s)
- Xiomara Q. Rosales
- Neuromuscular Center, Columbus, OH
- Department of Pediatrics, Neurology, and Center for Gene Therapy, Columbus, OH
- The Research Institute at Nationwide Children's Hospital, and The Ohio State University, Columbus, OH
| | - Julie M. Gastier-Foster
- Department of Pediatrics, Neurology, and Center for Gene Therapy, Columbus, OH
- The Research Institute at Nationwide Children's Hospital, and The Ohio State University, Columbus, OH
| | - Sarah Lewis
- Neuromuscular Center, Columbus, OH
- Department of Pediatrics, Neurology, and Center for Gene Therapy, Columbus, OH
| | - Malik Vinod
- Neuromuscular Center, Columbus, OH
- Department of Pediatrics, Neurology, and Center for Gene Therapy, Columbus, OH
| | - Devon L. Thrush
- Department of Pediatrics, Neurology, and Center for Gene Therapy, Columbus, OH
- The Research Institute at Nationwide Children's Hospital, and The Ohio State University, Columbus, OH
| | - Caroline Astbury
- Department of Pediatrics, Neurology, and Center for Gene Therapy, Columbus, OH
- The Research Institute at Nationwide Children's Hospital, and The Ohio State University, Columbus, OH
| | - Robert Pyatt
- Department of Pediatrics, Neurology, and Center for Gene Therapy, Columbus, OH
- The Research Institute at Nationwide Children's Hospital, and The Ohio State University, Columbus, OH
| | - Shalini Reshmi
- Department of Pediatrics, Neurology, and Center for Gene Therapy, Columbus, OH
- The Research Institute at Nationwide Children's Hospital, and The Ohio State University, Columbus, OH
| | - Zarife Sahenk
- Neuromuscular Center, Columbus, OH
- Department of Pediatrics, Neurology, and Center for Gene Therapy, Columbus, OH
- The Research Institute at Nationwide Children's Hospital, and The Ohio State University, Columbus, OH
| | - Jerry R. Mendell
- Neuromuscular Center, Columbus, OH
- Department of Pediatrics, Neurology, and Center for Gene Therapy, Columbus, OH
- The Research Institute at Nationwide Children's Hospital, and The Ohio State University, Columbus, OH
| |
Collapse
|
33
|
Glover LE, Newton K, Krishnan G, Bronson R, Boyle A, Krivickas LS, Brown RH. Dysferlin overexpression in skeletal muscle produces a progressive myopathy. Ann Neurol 2010; 67:384-93. [PMID: 20373350 DOI: 10.1002/ana.21926] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE The dose-response effects of dysferlin transgenesis were analyzed to determine if the dysferlin-deficient myopathies are good candidates for gene replacement therapy. METHODS We have generated 3 lines of transgenic mice, expressing low, mid, and high levels of full-length human dysferlin from a muscle-specific promoter. Transgenic skeletal muscle was analyzed and scored for morphological and functional deficits. RESULTS Overexpression of dysferlin in mice resulted in a striking phenotype of kyphosis, irregular gait, and reduced muscle mass and strength. Moreover, protein dosage correlated with phenotype severity. In contrast to dysferlin-null skeletal muscle, no evidence of sarcolemmal impairment was revealed. Rather, increased levels of Ca(2+)-regulated, dysferlin-binding proteins and endoplasmic reticulum stress chaperone proteins were observed in muscle lysates from transgenic mice as compared with controls. INTERPRETATION Expression levels of dysferlin are important for appropriate function without deleterious or cytotoxic effects. As a corollary, we propose that future endeavors in gene replacement for correction of dysferlinopathy should be tailored to take account of this.
Collapse
Affiliation(s)
- Louise E Glover
- Day Neuromuscular Research Laboratory, Massachusetts General Hospital, Charlestown, MA
| | | | | | | | | | | | | |
Collapse
|
34
|
Stuelsatz P, Pouzoulet F, Lamarre Y, Dargelos E, Poussard S, Leibovitch S, Cottin P, Veschambre P. Down-regulation of MyoD by calpain 3 promotes generation of reserve cells in C2C12 myoblasts. J Biol Chem 2010; 285:12670-83. [PMID: 20139084 PMCID: PMC2857084 DOI: 10.1074/jbc.m109.063966] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 01/25/2010] [Indexed: 01/24/2023] Open
Abstract
Calpain 3 is a calcium-dependent cysteine protease that is primarily expressed in skeletal muscle and is implicated in limb girdle muscular dystrophy type 2A. To date, its best characterized function is located within the sarcomere, but this protease is found in other cellular compartments, which suggests that it exerts multiple roles. Here, we present evidence that calpain 3 is involved in the myogenic differentiation process. In the course of in vitro culture of myoblasts to fully differentiated myotubes, a population of quiescent undifferentiated "reserve cells" are maintained. These reserve cells are closely related to satellite cells responsible for adult muscle regeneration. In the present work, we observe that reserve cells express higher levels of endogenous Capn3 mRNA than proliferating myoblasts. We show that calpain 3 participates in the establishment of the pool of reserve cells by decreasing the transcriptional activity of the key myogenic regulator MyoD via proteolysis independently of the ubiquitin-proteasome degradation pathway. Our results identify calpain 3 as a potential new player in the muscular regeneration process by promoting renewal of the satellite cell compartment.
Collapse
Affiliation(s)
- Pascal Stuelsatz
- From the
Université Bordeaux 1, Unité Protéolyse Croissance et Développement Musculaire, Institut National de la Recherche Agronomique (INRA), USC 2009, Avenue des Facultés, F-33405 Talence, France and
| | - Frédéric Pouzoulet
- From the
Université Bordeaux 1, Unité Protéolyse Croissance et Développement Musculaire, Institut National de la Recherche Agronomique (INRA), USC 2009, Avenue des Facultés, F-33405 Talence, France and
| | - Yann Lamarre
- From the
Université Bordeaux 1, Unité Protéolyse Croissance et Développement Musculaire, Institut National de la Recherche Agronomique (INRA), USC 2009, Avenue des Facultés, F-33405 Talence, France and
| | - Elise Dargelos
- From the
Université Bordeaux 1, Unité Protéolyse Croissance et Développement Musculaire, Institut National de la Recherche Agronomique (INRA), USC 2009, Avenue des Facultés, F-33405 Talence, France and
| | - Sylvie Poussard
- From the
Université Bordeaux 1, Unité Protéolyse Croissance et Développement Musculaire, Institut National de la Recherche Agronomique (INRA), USC 2009, Avenue des Facultés, F-33405 Talence, France and
| | - Serge Leibovitch
- the
Laboratoire de Génomique Fonctionnelle et Myogenèse, UMR866 Différenciation Cellulaire et Croissance, INRA UM II, Campus INRA/SupAgro, F-34060 Montpellier, France
| | - Patrick Cottin
- From the
Université Bordeaux 1, Unité Protéolyse Croissance et Développement Musculaire, Institut National de la Recherche Agronomique (INRA), USC 2009, Avenue des Facultés, F-33405 Talence, France and
| | - Philippe Veschambre
- From the
Université Bordeaux 1, Unité Protéolyse Croissance et Développement Musculaire, Institut National de la Recherche Agronomique (INRA), USC 2009, Avenue des Facultés, F-33405 Talence, France and
| |
Collapse
|
35
|
Azakir BA, Di Fulvio S, Therrien C, Sinnreich M. Dysferlin interacts with tubulin and microtubules in mouse skeletal muscle. PLoS One 2010; 5:e10122. [PMID: 20405035 PMCID: PMC2853571 DOI: 10.1371/journal.pone.0010122] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2009] [Accepted: 03/11/2010] [Indexed: 12/26/2022] Open
Abstract
Dysferlin is a type II transmembrane protein implicated in surface membrane repair in muscle. Mutations in dysferlin lead to limb girdle muscular dystrophy 2B, Miyoshi Myopathy and distal anterior compartment myopathy. Dysferlin's mode of action is not well understood and only a few protein binding partners have thus far been identified. Using affinity purification followed by liquid chromatography/mass spectrometry, we identified alpha-tubulin as a novel binding partner for dysferlin. The association between dysferlin and alpha-tubulin, as well as between dysferlin and microtubules, was confirmed in vitro by glutathione S-transferase pulldown and microtubule binding assays. These interactions were confirmed in vivo by co-immunoprecipitation. Confocal microscopy revealed that dysferlin and alpha-tubulin co-localized in the perinuclear region and in vesicular structures in myoblasts, and along thin longitudinal structures reminiscent of microtubules in myotubes. We mapped dysferlin's alpha-tubulin-binding region to its C2A and C2B domains. Modulation of calcium levels did not affect dysferlin binding to alpha-tubulin, suggesting that this interaction is calcium-independent. Our studies identified a new binding partner for dysferlin and suggest a role for microtubules in dysferlin trafficking to the sarcolemma.
Collapse
Affiliation(s)
- Bilal A. Azakir
- Neuromuscular Research Group, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Sabrina Di Fulvio
- Neuromuscular Research Group, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Christian Therrien
- Neuromuscular Research Group, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
| | - Michael Sinnreich
- Neuromuscular Research Group, Montreal Neurological Institute and Hospital, McGill University, Montreal, Quebec, Canada
- Neuromuscular Center, Departments of Neurology and Biomedicine, University Hospital Basel, Basel, Switzerland
- * E-mail:
| |
Collapse
|
36
|
Mellgren RL, Miyake K, Kramerova I, Spencer MJ, Bourg N, Bartoli M, Richard I, Greer PA, McNeil PL. Calcium-dependent plasma membrane repair requires m- or mu-calpain, but not calpain-3, the proteasome, or caspases. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2009; 1793:1886-93. [PMID: 19781581 DOI: 10.1016/j.bbamcr.2009.09.013] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2009] [Revised: 09/08/2009] [Accepted: 09/14/2009] [Indexed: 10/20/2022]
Abstract
Mechanically damaged plasma membrane undergoes rapid calcium-dependent resealing that appears to depend, at least in part, on calpain-mediated cortical cytoskeletal remodeling. Cells null for Capns1, the non-catalytic small subunit present in both m- and mu-calpains, do not undergo calcium-mediated resealing. However, it is not known which of these calpains is needed for repair, or whether other major cytosolic proteinases may participate. Utilizing isozyme-selective siRNAs to decrease expression of Capn1 or Capn2, catalytic subunits of mu- and m-calpains, respectively, in a mouse embryonic fibroblast cell line, we now show that substantial loss of both activities is required to compromise calcium-mediated survival after cell scrape-damage. Using skeletal myotubes derived from Capn3-null mice, we were unable to demonstrate loss of sarcolemma resealing after needle scratch or laser damage. Isolated muscle fibers from Capn3 knockout mice also efficiently repaired laser damage. Employing either a cell line expressing a temperature sensitive E1 ubiquitin ligase, or lactacystin, a specific proteasome inhibitor, it was not possible to demonstrate an effect of the proteasome on calcium-mediated survival after injury. Moreover, several cell-permeant caspase inhibitors were incapable of significantly decreasing survival or inhibiting membrane repair. Taken together with previous studies, the results show that m- or mu-calpain can facilitate repair of damaged plasma membrane. While there was no evidence for the involvement of calpain-3, the proteasome or caspases in early events of plasma membrane repair, our studies do not rule out their participation in downstream events that may link plasma membrane repair to adaptive remodeling after injury.
Collapse
Affiliation(s)
- Ronald L Mellgren
- Department of Physiology and Pharmacology, University of Toledo College of Medicine, Toledo, OH, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Wallace GQ, McNally EM. Mechanisms of muscle degeneration, regeneration, and repair in the muscular dystrophies. Annu Rev Physiol 2009; 71:37-57. [PMID: 18808326 DOI: 10.1146/annurev.physiol.010908.163216] [Citation(s) in RCA: 240] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
To withstand the rigors of contraction, muscle fibers have specialized protein complexes that buffer against mechanical stress and a multifaceted repair system that is rapidly activated after injury. Genetic studies first identified the mechanosensory signaling network that connects the structural elements of muscle and, more recently, have identified repair elements of muscle. Defects in the genes encoding the components of these systems lead to muscular dystrophy, a family of genetic disorders characterized by progressive muscle wasting. Although the age of onset, affected muscles, and severity vary considerably, all muscular dystrophies are characterized by muscle necrosis that overtakes the regenerative capacity of muscle. The resulting replacement of muscle by fatty and fibrous tissue leaves muscle increasingly weak and nonfunctional. This review discusses the cellular mechanisms that are primarily and secondarily disrupted in muscular dystrophy, focusing on membrane degeneration, muscle regeneration, and the repair of muscle.
Collapse
Affiliation(s)
- Gregory Q Wallace
- Department of Medicine, The University of Chicago, Chicago, Illinois 60637, USA
| | | |
Collapse
|
38
|
Beckmann JS, Spencer M. Calpain 3, the "gatekeeper" of proper sarcomere assembly, turnover and maintenance. Neuromuscul Disord 2008; 18:913-21. [PMID: 18974005 PMCID: PMC2614824 DOI: 10.1016/j.nmd.2008.08.005] [Citation(s) in RCA: 105] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 08/18/2008] [Accepted: 08/27/2008] [Indexed: 11/17/2022]
Abstract
Calpain 3 is a member of the calpain family of calcium-dependent intracellular proteases. Thirteen years ago it was discovered that mutations in calpain 3 (CAPN3) result in an autosomal recessive and progressive form of limb girdle muscular dystrophy called limb girdle muscular dystrophy type 2A. While calpain 3 mRNA is expressed at high levels in muscle and appears to have some role in developmental processes, muscles of patients and mice lacking calpain 3 still form apparently normal muscle during prenatal development; thus, a functional calpain 3 protease is not mandatory for muscle to form in vivo but it is a pre-requisite for muscle to remain healthy. Despite intensive research in this field, the physiological substrates of the calpain 3 protein (hereafter referred to as CAPN3) and its alternatively spliced isoforms remain elusive. The existence of these multiple isoforms complicates the search for the physiological functions of CAPN3 and its pathophysiological role. In this review, we summarize the genetic and biochemical evidence that point to loss of function of the full-length isoform of CAPN3, also known as p94, as the pathogenic isoform. We also argue that its natural substrates must reside in its proximity within the sarcomere where it is stored in an inactive state anchored to titin. We further propose that CAPN3 has many attributes that make it ideally suited as a sensor of sarcomeric integrity and function, involved in its repair and maintenance. Loss of these CAPN3-mediated activities can explain the "progressive" development of muscular dystrophy.
Collapse
Affiliation(s)
- Jacques S Beckmann
- Service and Department of Medical Genetics, Centre Hospitalier Universitaire Vaudois, CHUV and Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland.
| | | |
Collapse
|
39
|
Sáenz A, Azpitarte M, Armañanzas R, Leturcq F, Alzualde A, Inza I, García-Bragado F, De la Herran G, Corcuera J, Cabello A, Navarro C, De la Torre C, Gallardo E, Illa I, López de Munain A. Gene expression profiling in limb-girdle muscular dystrophy 2A. PLoS One 2008; 3:e3750. [PMID: 19015733 PMCID: PMC2582180 DOI: 10.1371/journal.pone.0003750] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2008] [Accepted: 10/25/2008] [Indexed: 11/18/2022] Open
Abstract
Limb-girdle muscular dystrophy type 2A (LGMD2A) is a recessive genetic disorder caused by mutations in calpain 3 (CAPN3). Calpain 3 plays different roles in muscular cells, but little is known about its functions or in vivo substrates. The aim of this study was to identify the genes showing an altered expression in LGMD2A patients and the possible pathways they are implicated in. Ten muscle samples from LGMD2A patients with in which molecular diagnosis was ascertained were investigated using array technology to analyze gene expression profiling as compared to ten normal muscle samples. Upregulated genes were mostly those related to extracellular matrix (different collagens), cell adhesion (fibronectin), muscle development (myosins and melusin) and signal transduction. It is therefore suggested that different proteins located or participating in the costameric region are implicated in processes regulated by calpain 3 during skeletal muscle development. Genes participating in the ubiquitin proteasome degradation pathway were found to be deregulated in LGMD2A patients, suggesting that regulation of this pathway may be under the control of calpain 3 activity. As frizzled-related protein (FRZB) is upregulated in LGMD2A muscle samples, it could be hypothesized that beta-catenin regulation is also altered at the Wnt signaling pathway, leading to an incorrect myogenesis. Conversely, expression of most transcription factor genes was downregulated (MYC, FOS and EGR1). Finally, the upregulation of IL-32 and immunoglobulin genes may induce the eosinophil chemoattraction explaining the inflammatory findings observed in presymptomatic stages. The obtained results try to shed some light on identification of novel therapeutic targets for limb-girdle muscular dystrophies.
Collapse
Affiliation(s)
- Amets Sáenz
- Experimental Unit, Hospital Donostia, Donostia-San Sebastián, Basque Country, Spain.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Kramerova I, Kudryashova E, Wu B, Ottenheijm C, Granzier H, Spencer MJ. Novel role of calpain-3 in the triad-associated protein complex regulating calcium release in skeletal muscle. Hum Mol Genet 2008; 17:3271-80. [PMID: 18676612 PMCID: PMC2566524 DOI: 10.1093/hmg/ddn223] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2008] [Accepted: 07/30/2008] [Indexed: 02/02/2023] Open
Abstract
Calpain-3 (CAPN3) is a non-lysosomal cysteine protease that is necessary for normal muscle function, as mutations in CAPN3 result in an autosomal recessive form of limb girdle muscular dystrophy type 2A. To elucidate the biological roles of CAPN3 in skeletal muscle, we performed a search for potential substrates and interacting partners. By yeast-two-hybrid analysis we identified the glycolytic enzyme aldolase A (AldoA) as a binding partner of CAPN3. In co-expression studies CAPN3 degraded AldoA; however, no accumulation of AldoA was observed in total extracts from CAPN3-deficient muscles suggesting that AldoA is not an in vivo substrate of CAPN3. Instead, we found CAPN3 to be necessary for recruitment of AldoA to one specific location, namely the triads, which are structural components of muscle responsible for calcium transport and excitation-contraction coupling. Both aldolase and CAPN3 are present in the triad-enriched fraction and are able to interact with ryanodine receptors (RyR) that form major calcium release channels. Levels of triad-associated AldoA and RyR were decreased in CAPN3-deficient muscles compared with wild-type. Consistent with these observations we found calcium release to be significantly reduced in fibers from CAPN3-deficient muscles. Together, these data suggest that CAPN3 is necessary for the structural integrity of the triad-associated protein complex and that impairment of calcium transport is a phenotypic feature of CAPN3-deficient muscle.
Collapse
Affiliation(s)
- Irina Kramerova
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Elena Kudryashova
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Benjamin Wu
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| | - Coen Ottenheijm
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85724, USA
| | - Henk Granzier
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, AZ 85724, USA
| | - Melissa J. Spencer
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
41
|
Huang Y, de Morrée A, van Remoortere A, Bushby K, Frants RR, Dunnen JT, van der Maarel SM. Calpain 3 is a modulator of the dysferlin protein complex in skeletal muscle. Hum Mol Genet 2008; 17:1855-66. [PMID: 18334579 PMCID: PMC2900895 DOI: 10.1093/hmg/ddn081] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Accepted: 03/09/2008] [Indexed: 11/13/2022] Open
Abstract
Muscular dystrophies comprise a genetically heterogeneous group of degenerative muscle disorders characterized by progressive muscle wasting and weakness. Two forms of limb-girdle muscular dystrophy, 2A and 2B, are caused by mutations in calpain 3 (CAPN3) and dysferlin (DYSF), respectively. While CAPN3 may be involved in sarcomere remodeling, DYSF is proposed to play a role in membrane repair. The coexistence of CAPN3 and AHNAK, a protein involved in subsarcolemmal cytoarchitecture and membrane repair, in the dysferlin protein complex and the presence of proteolytic cleavage fragments of AHNAK in skeletal muscle led us to investigate whether AHNAK can act as substrate for CAPN3. We here demonstrate that AHNAK is cleaved by CAPN3 and show that AHNAK is lost in cells expressing active CAPN3. Conversely, AHNAK accumulates when calpain 3 is defective in skeletal muscle of calpainopathy patients. Moreover, we demonstrate that AHNAK fragments cleaved by CAPN3 have lost their affinity for dysferlin. Thus, our findings suggest interconnectivity between both diseases by revealing a novel physiological role for CAPN3 in regulating the dysferlin protein complex.
Collapse
Affiliation(s)
| | | | | | - Kate Bushby
- Institute of Human Genetics, International Centre for Life, Newcastle-upon-Tyne, UK
| | | | | | | |
Collapse
|
42
|
Spuler S, Carl M, Zabojszcza J, Straub V, Bushby K, Moore SA, Bähring S, Wenzel K, Vinkemeier U, Rocken C. Dysferlin-deficient muscular dystrophy features amyloidosis. Ann Neurol 2008; 63:323-8. [PMID: 18306167 DOI: 10.1002/ana.21309] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
OBJECTIVE Dysferlin (DYSF) gene mutations cause limb girdle muscular dystrophy type 2B and Miyoshi's myopathy. The consequences of DYSF mutations on protein structure are poorly understood. METHODS The gene encoding dysferlin was sequenced in patients with suspected dysferlin-deficient muscular dystrophy. Muscle biopsy specimens were analyzed by histochemistry, immunohistochemistry, and electron microscopy. Antibodies against N-terminal dysferlin-peptides were raised. RESULTS We found three families with muscular dystrophy caused by homozygous or compound heterozygous DYSF mutations featuring sarcolemmal and interstitial amyloid deposits. These mutations were all located in the N-terminal region of the protein. Dysferlin was a constituent of the amyloid deposits. INTERPRETATION Limb girdle muscular dystrophy type 2B is the first muscular dystrophy associated with amyloidosis. Molecular treatment strategies will necessarily have to consider the presence of amyloidogenesis.
Collapse
Affiliation(s)
- Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center at the Charité and the Max-Delbrück Center, Berlin, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Dargelos E, Poussard S, Brulé C, Daury L, Cottin P. Calcium-dependent proteolytic system and muscle dysfunctions: a possible role of calpains in sarcopenia. Biochimie 2007; 90:359-68. [PMID: 17881114 DOI: 10.1016/j.biochi.2007.07.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2007] [Accepted: 07/20/2007] [Indexed: 02/02/2023]
Abstract
The calcium-dependent proteolytic system is composed of cysteine proteases named calpains. They are ubiquitous or tissue-specific enzymes. The two best characterised isoforms are the ubiquitously expressed mu- and m-calpains. Besides its regulation by calcium, calpain activity is tightly controlled by calpastatin, the specific endogenous inhibitor, binding to phospholipids, autoproteolysis and phosphorylation. Calpains are responsible for limited proteolytic events. Among the multitude of substrates identified so far are cytoskeletal and membrane proteins, enzymes and transcription factors. Calpain activity is involved in a large number of physiological and pathological processes. In this review, we will particularly focus on the implication of the calcium-dependent proteolytic system in relation to muscle physiology. Because of their ability to remodel cytoskeletal anchorage complexes, calpains play a major role in the regulation of cell adhesion, migration and fusion, three key steps of myogenesis. Calcium-dependent proteolysis is also involved in the control of cell cycle. In muscle tissue, in particular, calpains intervene in the regeneration process. Another important class of calpain substrates belongs to apoptosis regulating factors. The proteases may thus play a role in muscle cell death, and as a consequence in muscle atrophy. The relationships between calcium-dependent proteolysis and muscle dysfunctions are being further developed in this review with a particular emphasis on sarcopenia.
Collapse
Affiliation(s)
- E Dargelos
- Université Bordeaux I, INRA USC-2009, Unité Protéolyse Croissance et Développement Musculaire, ISTAB, avenue des facultés, 33405 Talence cedex, France.
| | | | | | | | | |
Collapse
|
44
|
Abstract
The muscular dystrophies are a heterogeneous group of inherited disorders, defined by progressive muscle weakness and atrophy. Following the discovery of dystrophin, remarkable progress has been made in defining the molecular properties of proteins involved in the various dystrophies. This has underlined the importance of the dystrophin-associated protein complex as a cell membrane scaffold, providing structural stability to muscle cells (McNeil PL, Khakee R. Disruptions of muscle fiber plasma membranes. Role in exercise-induced damage. Am J Pathol 1992;140:1097-1109). While the dystrophies linked to loss of function of dystrophin and its associated proteins are caused by diminished membrane integrity, it is now believed that a new class of dystrophies arises because of a diminished capacity for rapid muscle membrane repair after injury. Dysferlin is the first identified member of a putative muscle-specific repair complex that permits rapid resealing of membranes disrupted by mechanical stress. Membrane resealing is a function conserved by most cells and is mediated by a mechanism closely resembling regulated, Ca2+-dependent exocytosis. A primary role for dysferlin in this pathway, as a Ca2+-regulated fusogen, has been suggested, and a number of candidate partner proteins have been identified. This review outlines the current understanding of the role of dysferlin in membrane repair and the evolving picture of dysferlin-related signaling pathways in muscle cell physiology and pathology.
Collapse
Affiliation(s)
- Louise Glover
- Day Neuromuscular Research Laboratory, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA 02129, USA
| | | |
Collapse
|
45
|
Klinge L, Laval S, Keers S, Haldane F, Straub V, Barresi R, Bushby K. From T-tubule to sarcolemma: damage-induced dysferlin translocation in early myogenesis. FASEB J 2007; 21:1768-76. [PMID: 17363620 DOI: 10.1096/fj.06-7659com] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The dysferlin gene is mutated in limb-girdle muscular dystrophy type 2B, Miyoshi myopathy, and distal anterior compartment myopathy. In mature skeletal muscle, dysferlin is located predominantly at the sarcolemma, where it plays a role in membrane fusion and repair. To investigate the role of dysferlin during early muscle differentiation, its localization was studied at high resolution in a muscle cell line. This demonstrated that dysferlin is not expressed at the plasmalemma of myotubes but mostly localizes to the T-tubule network. However, dysferlin translocated to the site of injury and toward the plasma membrane in a Ca2+-dependent fashion in response to a newly designed in vitro wounding assay. This reaction was specific to the full-length protein, as heterologously expressed deletion mutants of distinct C2 domains of dysferlin did not show this response. These results shed light on the dynamics of muscle membrane repair and are highly indicative of a specific role of dysferlin in this process in early myogenesis.
Collapse
Affiliation(s)
- Lars Klinge
- Institute of Human Genetics, University of Newcastle upon Tyne, International Centre for Life, Central Pkwy, NE1 3BZ Newcastle upon Tyne, England, UK
| | | | | | | | | | | | | |
Collapse
|
46
|
Milic A, Daniele N, Lochmüller H, Mora M, Comi GP, Moggio M, Noulet F, Walter MC, Morandi L, Poupiot J, Roudaut C, Bittner RE, Bartoli M, Richard I. A third of LGMD2A biopsies have normal calpain 3 proteolytic activity as determined by an in vitro assay. Neuromuscul Disord 2007; 17:148-56. [PMID: 17236769 DOI: 10.1016/j.nmd.2006.11.001] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 11/06/2006] [Accepted: 11/21/2006] [Indexed: 10/23/2022]
Abstract
Limb-girdle muscular dystrophy type 2A (LGMD2A) is an autosomal recessive muscular disorder caused by mutations in the gene coding for calpain 3, a calcium-dependent protease. We developed an in vitro assay that can detect the proteolytic activity of calpain 3 in a muscle sample. This assay is based on the use of an inactive calpain 3 as a substrate for active calpain 3 molecules. A total of 79 human biopsies have been analysed using an unbiased single blind method. Results were confronted with the molecular diagnosis for confirmation. Proteolytic activity was either reduced or absent in 68% of LGMD2A biopsies. In the remaining 32%, normal proteolytic activity was found despite the presence of calpain 3 mutation(s), suggesting that other calpain 3 properties might be impaired to give rise to the LGMD2A phenotype. Our assay is easily adaptable to routine and appears to be more sensitive than common analysis by immunodetection.
Collapse
Affiliation(s)
- Astrid Milic
- Laboratory of Clinical Neurogenetics and Muscular Disorders, Zagreb University School of Medicine, Croatian Institute for Brain Research, Salata 12, 10000 Zagreb, Croatia
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Huang Y, Laval SH, van Remoortere A, Baudier J, Benaud C, Anderson LVB, Straub V, Deelder A, Frants RR, den Dunnen JT, Bushby K, van der Maarel SM. AHNAK, a novel component of the dysferlin protein complex, redistributes to the cytoplasm with dysferlin during skeletal muscle regeneration. FASEB J 2006; 21:732-42. [PMID: 17185750 DOI: 10.1096/fj.06-6628com] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Mutations in dysferlin cause limb girdle muscular dystrophy 2B, Miyoshi myopathy and distal anterior compartment myopathy. Dysferlin is proposed to play a role in muscle membrane repair. To gain functional insight into the molecular mechanisms of dysferlin, we have searched for dysferlin-interacting proteins in skeletal muscle. By coimmunoprecipitation coupled with mass spectrometry, we demonstrate that AHNAK interacts with dysferlin. We defined the binding sites in dysferlin and AHNAK as the C2A domain in dysferlin and the carboxyterminal domain of AHNAK by glutathione S-transferase (GST)-pull down assays. As expected, the N-terminal domain of myoferlin also interacts with the carboxyterminal domain of AHNAK. In normal skeletal muscle, dysferlin and AHNAK colocalize at the sarcolemmal membrane and T-tubules. In dysferlinopathies, reduction or absence of dysferlin correlates with a secondary muscle-specific loss of AHNAK. Moreover, in regenerating rat muscle, dysferlin and AHNAK showed a marked increase and cytoplasmic localization, consistent with the direct interaction between them. Our data suggest that dysferlin participates in the recruitment and stabilization of AHNAK to the sarcolemma and that AHNAK plays a role in dysferlin membrane repair process. It may also have significant implications for understanding the biology of AHNAK-containing exocytotic vesicles, "enlargosomes," in plasma membrane remodeling and repair.
Collapse
Affiliation(s)
- Yanchao Huang
- Center for Human and Clinical Genetics, Leiden Univesity Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Cohen N, Kudryashova E, Kramerova I, Anderson LVB, Beckmann JS, Bushby K, Spencer MJ. Identification of putative in vivo substrates of calpain 3 by comparative proteomics of overexpressing transgenic and nontransgenic mice. Proteomics 2006; 6:6075-84. [PMID: 17051641 DOI: 10.1002/pmic.200600199] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Calpain 3 (CAPN3) is a calcium-dependent protease, mutations in which cause limb girdle muscular dystrophy type 2A. To explore the physiological function of CAPN3, we compared the proteomes of transgenic mice that overexpress CAPN3 (CAPN3 Tg) and their nontransgenic (non-Tg) counterparts. We first examined known muscular dystrophy-related proteins to determine if overexpression of CAPN3 results in a change in their distribution or concentration. This analysis did not identify any known muscular dystrophy proteins as substrates of CAPN3. Next, we used a proteomic approach to compare and identify differentially represented proteins in 2-DE of CAPN3 Tg and non-Tg mice. LC-MS/MS analysis led to the identification of ten possible substrates for CAPN3, classified into two major functional categories: metabolic and myofibrillar. Myosin light chain 1 (MLC1) was focused upon because our previous studies suggested a role for CAPN3 in sarcomere remodeling. In this study, CAPN3 was shown to proteolyze MLC1 in vitro. These studies are the first to identify possible substrates for CAPN3 in an in vivo system and support a role for CAPN3 in sarcomere remodeling by cleavage of myofibrillar proteins such as MLC1. In addition, these data also suggest a role for CAPN3 in mitochondrial protein turnover.
Collapse
Affiliation(s)
- Niaz Cohen
- Department of Neurology, and UCLA Duchenne Muscular Dystrophy Research Center, University of California, Los Angeles, CA 90095-7334, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Abstract
Calpain 3 is a 94-kDa calcium-dependent cysteine protease mainly expressed in skeletal muscle. In this tissue, it localizes at several regions of the sarcomere through binding to the giant protein, titin. Loss-of-function mutations in the calpain 3 gene have been associated with limb-girdle muscular dystrophy type 2A (LGMD2A), a common form of muscular dystrophy found world wide. Recently, significant progress has been made in understanding the mode of regulation and the possible function of calpain 3 in muscle. It is now well accepted that it has an unusual zymogenic activation and that cytoskeletal proteins are one class of its substrates. Through the absence of cleavage of these substrates, calpain 3 deficiency leads to abnormal sarcomeres, impairment of muscle contractile capacity, and death of the muscle fibers. These data indicate a role for calpain 3 as a chef d'orchestre in sarcomere remodeling and suggest a new category of LGMD2 pathological mechanisms.
Collapse
|
50
|
Kramerova I, Beckmann JS, Spencer MJ. Molecular and cellular basis of calpainopathy (limb girdle muscular dystrophy type 2A). Biochim Biophys Acta Mol Basis Dis 2006; 1772:128-44. [PMID: 16934440 DOI: 10.1016/j.bbadis.2006.07.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2006] [Revised: 07/07/2006] [Accepted: 07/13/2006] [Indexed: 11/28/2022]
Abstract
Limb girdle muscular dystrophy type 2A results from mutations in the gene encoding the calpain 3 protease. Mutations in this disease are inherited in an autosomal recessive fashion and result in progressive proximal skeletal muscle wasting but no cardiac abnormalities. Calpain 3 has been shown to proteolytically cleave a wide variety of cytoskeletal and myofibrillar proteins and to act upstream of the ubiquitin-proteasome pathway. In this review, we summarize the known biochemical and physiological features of calpain 3 and hypothesize why mutations result in disease.
Collapse
Affiliation(s)
- Irina Kramerova
- Department of Neurology and Pediatrics and UCLA Duchenne Muscular Dystrophy Research Center, University of California, Los Angeles, Neuroscience Research Building, 635 Young Dr. South, Los Angeles, CA 90095-7334, USA
| | | | | |
Collapse
|