1
|
Poudel BH, Fletcher S, Wilton SD, Aung-Htut M. Limb Girdle Muscular Dystrophy Type 2B (LGMD2B): Diagnosis and Therapeutic Possibilities. Int J Mol Sci 2024; 25:5572. [PMID: 38891760 PMCID: PMC11171558 DOI: 10.3390/ijms25115572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 05/11/2024] [Accepted: 05/16/2024] [Indexed: 06/21/2024] Open
Abstract
Dysferlin is a large transmembrane protein involved in critical cellular processes including membrane repair and vesicle fusion. Mutations in the dysferlin gene (DYSF) can result in rare forms of muscular dystrophy; Miyoshi myopathy; limb girdle muscular dystrophy type 2B (LGMD2B); and distal myopathy. These conditions are collectively known as dysferlinopathies and are caused by more than 600 mutations that have been identified across the DYSF gene to date. In this review, we discuss the key molecular and clinical features of LGMD2B, the causative gene DYSF, and the associated dysferlin protein structure. We also provide an update on current approaches to LGMD2B diagnosis and advances in drug development, including splice switching antisense oligonucleotides. We give a brief update on clinical trials involving adeno-associated viral gene therapy and the current progress on CRISPR/Cas9 mediated therapy for LGMD2B, and then conclude by discussing the prospects of antisense oligomer-based intervention to treat selected mutations causing dysferlinopathies.
Collapse
Affiliation(s)
- Bal Hari Poudel
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Perth, WA 6009, Australia
- Central Department of Biotechnology, Tribhuvan University, Kirtipur, Kathmandu 44618, Nepal
| | - Sue Fletcher
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
| | - Steve D. Wilton
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Perth, WA 6009, Australia
| | - May Aung-Htut
- Centre for Molecular Medicine and Innovative Therapeutics, Health Futures Institute, Murdoch University, Perth, WA 6150, Australia; (B.H.P.); (S.F.); (S.D.W.)
- Perron Institute for Neurological and Translational Science, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
2
|
Zhou J, Zhou R, Feng Q, Song X, Chen X. Phenotypic and genotypic analysis of a patient with Miyoshi myopathy caused by truncated protein. Gene 2024; 893:147929. [PMID: 38381504 DOI: 10.1016/j.gene.2023.147929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2023] [Revised: 10/16/2023] [Accepted: 10/25/2023] [Indexed: 02/22/2024]
Abstract
Dysferlin protein deficiency can cause neuromuscular dysfunction, resulting in autosomal recessive dysferlinopathy, which is caused by DYSF gene mutation. Dysferlin proteins belongs to the Ferlin1-like protein family and are associated with muscle membrane repair and regeneration. In China, pathogenic mutations of the protein often result in two clinical phenotypes of Miyoshi muscular or limb band muscular dystrophy type 2B. It is clinically characterized by progressive muscle weakness and elevated serum creatine kinase. The data of the child were collected, blood samples of the child and his family members were collected, and whole exome sequencing (WES) was performed. The recombinant expression vector was constructed, the function of the mutation was verified by minigene, and the pathogenicity of the mutation was further analyzed by combining with biological information analysis. The patient initially presented with asymptomatic elevation of serum creatine kinase(CK). Then progressive lower limb weakness, mainly distal limb weakness. Large amounts of scattered necrosis, myogenic lesions, and complete deletion of dysferlin protein were observed under muscle biopsy, which further improved genetic detection. Whole exome sequencing showed compound mutations (c.1397 + 1_1397 + 3del and c.1375dup p.M459Nfs*15) in DYSF gene. c.1375dup p.M459Nfs*15 have been reported. The other mutation is the deletion of c.1397 + 1_1397 + 3 in Intron15, which is an intron mutation that may affect splicing and the pathogenesis is still unknown. Minigene splicing assay verified that c.1397 + 1_1397 + 3del resulted in exon15 skipping and produced a premature termination codon. We report a novel pathogenic mutation in DYSF gene with Miyoshi myopathy and demonstrate this variant causes skipping of exon15 by minigene splicing assay. We point out the need of conducting functional analysis to verify the pathogenicity of intronic mutation. The finding enriches the mutation spectrum of DYSF gene and laid a foundation for future studies on the correlation between genotype and phenotype.
Collapse
Affiliation(s)
- Jinyi Zhou
- Children's Hospital of Soochow University, Department of Neurology, China
| | - Rui Zhou
- Children's Hospital of Soochow University, Department of Neurology, China
| | - Qihua Feng
- Children's Hospital of Soochow University, Department of Rheumatology and Immunology, China
| | - Xiaoxiang Song
- Children's Hospital of Soochow University, Department of Rheumatology and Immunology, China
| | - Xuqin Chen
- Children's Hospital of Soochow University, Department of Neurology, China.
| |
Collapse
|
3
|
Chen Y, Wu W, Wang P, Yip P, Wu Y, Lin Y, Lin W. Novel five nucleotide deletion in dysferlin leads to autosomal recessive limb-girdle muscular dystrophy. Physiol Rep 2023; 11:e15887. [PMID: 38110300 PMCID: PMC10727958 DOI: 10.14814/phy2.15887] [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: 07/12/2023] [Revised: 12/01/2023] [Accepted: 12/02/2023] [Indexed: 12/20/2023] Open
Abstract
Muscular dystrophy (MD) is a genetic disorder that causes progressive muscle weakness and degeneration. Limb-girdle muscular dystrophy (LGMD) is a type of MD that mainly causes muscle atrophy within the shoulder and pelvic girdles. LGMD is classified into autosomal dominant (LGMD-D) and autosomal recessive (LGMD-R) inheritance patterns. Mutations in the Dysferlin gene (DYSF) are common causes of LGMD-R. However, genetic screening of DYSF mutations is rare in Taiwan. Herein, we identified a novel c.2867_2871del ACCAG deletion and a previously reported c.937+1G>A mutation in DYSF from a Taiwanese family with LGMD. The primary symptoms of both siblings were difficulty climbing stairs, walking on the toes, and gradually worsening weakness in the proximal muscles and increased creatine kinase level. Through pedigree analysis and sequencing, two siblings from this family were found to have compound heterozygous DYSF mutations (c. 937+1G>A and c. 2867_2871del ACCAG) within the separated alleles. These mutations induced early stop codons; if translated, truncated DYSF proteins will be expressed. Or, the mRNA products of these two mutations will merit the nonsense-mediated decay, might result in no dysferlin protein expressed. To our knowledge, this is the first report of a novel c.2867_2871del ACCAG deletion in DYSF. Further research is required to examine the effects of the novel DYSF mutation in Taiwanese patients with LGMD.
Collapse
Affiliation(s)
- Yen‐Lin Chen
- Center for Precision Medicine and Genomics, Tri‐Service General HospitalMedical Defense Medical CenterTaipeiTaiwan
- Department of Pathology, Tri‐Service General HospitalMedical Defense Medical CenterTaipeiTaiwan
| | - Wen‐Bin Wu
- School of Medicine, College of MedicineFu Je Catholic UniversityNew Taipei CityTaiwan
| | - Pei Wang
- School of Medicine, College of MedicineFu Je Catholic UniversityNew Taipei CityTaiwan
| | - Ping‐Keung Yip
- School of Medicine, College of MedicineFu Je Catholic UniversityNew Taipei CityTaiwan
- Division of NeurologyCardinal Tien HospitalNew Taipei CityTaiwan
| | - Yi‐No Wu
- School of Medicine, College of MedicineFu Je Catholic UniversityNew Taipei CityTaiwan
| | - Ying‐Hung Lin
- Graduate Institute of Biomedical and Pharmaceutical ScienceFu Jen Catholic UniversityNew Taipei CityTaiwan
| | - Wei‐Ning Lin
- Graduate Institute of Biomedical and Pharmaceutical ScienceFu Jen Catholic UniversityNew Taipei CityTaiwan
| |
Collapse
|
4
|
Bardakov SN, Deev RV, Isaev АА, Khromov‐Borisov NN, Kopylov ED, Savchuk МR, Pushkin MS, Presnyakov EV, Magomedova RM, Achmedova PG, Umakhanova ZR, Kaimonov VS, Musatova EV, Blagodatskikh KА, Tveleneva AА, Sofronova YV, Yakovlev IA. Genetic screening of an endemic mutation in the DYSF gene in an isolated, mountainous population in the Republic of Dagestan. Mol Genet Genomic Med 2023; 11:e2236. [PMID: 37553796 PMCID: PMC10568376 DOI: 10.1002/mgg3.2236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 06/24/2023] [Accepted: 06/29/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Dysferlinopathy has a high prevalence in relatively isolated ethnic groups where consanguineous marriages are characteristic and/or the founder effect exists. However, the frequency of endemic mutations in most isolates has not been investigated. METHODS The prevalence of the pathological DYSF gene variant (NM_003494.4); c.200_201delinsAT, p. Val67Asp (rs121908957) was investigated in an isolated Avar population in the Republic of Dagestan. Genetic screenings were conducted in a remote mountainous region characterized by a high level of consanguinity among its inhabitants. In total, 746 individuals were included in the screenings. RESULTS This pathological DYSF gene variant causes two primary phenotypes of dysferlinopathy: limb-girdle muscular dystrophy (LGMD) type R2 and Miyoshi muscular dystrophy type 1. Results indicated a high prevalence of the allele at 14% (95% confidence interval [CI]: 12-17; 138 out of 1518 alleles), while the allele in the homozygous state was detected in 29 cases-3.8% (CI: 2.6-5.4). The population load for dysferlinopathy was 832.3 ± 153.9 per 100,000 with an average prevalence of limb-girdle muscular dystrophies ranging from 0.38 ± 0.38 to 5.93 ± 1.44 per 100,000. CONCLUSION A significant burden of the allele was due to inbreeding, as evidenced by a deficiency of heterozygotes and the Wright fixation index equal to 0.14 (CI 0.06-0.23).
Collapse
Affiliation(s)
| | - Roman V. Deev
- North‐Western State Medical University named after I.I. MechnikovSaint PetersburgRussia
- Human Stem Cells InstituteMoscowRussia
| | - Аrtur А. Isaev
- Human Stem Cells InstituteMoscowRussia
- Genotarget LLCSkolkovo Innovation CentreMoscowRussia
| | | | - Evgeniy D. Kopylov
- North‐Western State Medical University named after I.I. MechnikovSaint PetersburgRussia
| | - Мaria R. Savchuk
- North‐Western State Medical University named after I.I. MechnikovSaint PetersburgRussia
| | - Maxim S. Pushkin
- North‐Western State Medical University named after I.I. MechnikovSaint PetersburgRussia
| | - Evgeniy V. Presnyakov
- North‐Western State Medical University named after I.I. MechnikovSaint PetersburgRussia
| | | | | | | | | | | | | | | | | | - Ivan A. Yakovlev
- Human Stem Cells InstituteMoscowRussia
- Genotarget LLCSkolkovo Innovation CentreMoscowRussia
| |
Collapse
|
5
|
Akbar F, Saleem SM, Khalid E, Ibrahim S, Afroze B, Kirmani S, Khan S. The spectrum of hereditary neuromuscular disorders in the Pakistani population. Am J Med Genet A 2023; 191:2536-2550. [PMID: 37366078 DOI: 10.1002/ajmg.a.63332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 05/21/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023]
Abstract
Hereditary neuromuscular disorders (NMDs) are a broad group of clinically heterogeneous disorders with varying inheritance patterns, that are associated with over 500 implicated genes. In the context of a highly consanguineous Pakistani population, we expect that autosomal recessive NMDs may have a higher prevalence compared with patients of European descent. This is the first study to offer a detailed description of the spectrum of genes causing hereditary NMDs in the Pakistani population using NGS testing. To study the clinical and genetic profiles of patients presenting for evaluation of a hereditary neuromuscular disorder. This is a retrospective chart review of patients seen in the Neuromuscular Disorders Clinic and referred to the Genetics Clinic with a suspected hereditary neuromuscular disorder, between 2016 and 2020 at the Aga Khan University Hospital, Karachi and Mukhtiar A. Sheikh Hospital, Multan, Pakistan. The genetic testing for these patients included NGS-based single gene sequencing, NGS-based multi-gene panel and whole exome sequencing. In a total of 112 patients studied, 35 (31.3%) were female. The mean age of onset in all patients was 14.6 years (SD ±12.1 years), with the average age at presentation to the clinic of 22.4 years (SD ±14.10 years). Forty-seven (41.9%) patients had a positive genetic test result, 53 (47.3%) had one or more variants of uncertain significance (VUS), and 12 (10.7%) had a negative result. Upon further genotype-phenotype correlation and family segregation analysis, the diagnostic yield improved, with 59 (52.7%) patients reaching a diagnosis of a hereditary NMD. We also report probable founder variants in COL6A2, FKTN, GNE, and SGCB, previously reported in populations that have possible shared ancestry with the Pakistani population. Our findings reemphasizes that the rate of VUSs can be reduced by clinical correlation and family segregation studies.
Collapse
Affiliation(s)
- Fizza Akbar
- Division of Women and Child Health, The Aga Khan University, Karachi, Pakistan
| | | | | | - Shahnaz Ibrahim
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Bushra Afroze
- Department of Paediatrics and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Salman Kirmani
- Division of Women and Child Health, The Aga Khan University, Karachi, Pakistan
| | - Sara Khan
- Department of Neurology, The Aga Khan University, Karachi, Pakistan
| |
Collapse
|
6
|
Sun C, Xie Z, Cong L, Xu Y, Liu Z. An in-frame pseudoexon activation caused by a novel deep-intronic variant in the dysferlin gene. Ann Clin Transl Neurol 2023; 10:292-296. [PMID: 36542547 PMCID: PMC9930419 DOI: 10.1002/acn3.51716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/23/2022] [Accepted: 11/23/2022] [Indexed: 12/24/2022] Open
Abstract
The precise detection and interpretation of pathogenic DYSF variants are sometimes challenging, largely due to rare deep-intronic splice-altering variants. Here, we report on the genetic diagnosis of a male patient with dysferlinopathy. He remained genetically unsolved after routine exonic detection approaches that only detected a novel heterozygous frameshift variant (c.407dup, p.Thr137Tyrfs*11) in DYSF exon 5. Via muscle-derived DYSF mRNA studies, we identified a novel deep-intronic DYSF variant in the other allele (c.1397 + 649C > T), which causing in-frame alterations in DYSF mRNA and protein structure and confirmed his genetic diagnosis of dysferlinopathy. Our study emphasizes the potential role of undetected deep-intronic splice-altering variants in monogenic diseases.
Collapse
Affiliation(s)
- Chengyue Sun
- Department of Neurology, Peking University People's Hospital, Beijing, 100044, China
| | - Zhiying Xie
- Department of Neurology, Peking University First Hospital, Beijing, 100034, China
| | - Lu Cong
- Department of Neurology, Peking University People's Hospital, Beijing, 100044, China
| | - Yan Xu
- Department of Neurology, Peking University People's Hospital, Beijing, 100044, China
| | - Zunjing Liu
- Department of Neurology, Peking University People's Hospital, Beijing, 100044, China
| |
Collapse
|
7
|
Carpenter AP, Khuu P, Weidner T, Johnson CP, Roeters SJ, Baio JE. Orientation of the Dysferlin C2A Domain is Responsive to the Composition of Lipid Membranes. J Phys Chem B 2023; 127:577-589. [PMID: 36608331 DOI: 10.1021/acs.jpcb.2c06716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Dysferlin is a 230 kD protein that plays a critical function in the active resealing of micron-sized injuries to the muscle sarcolemma by recruiting vesicles to patch the injured site via vesicle fusion. Muscular dystrophy is observed in humans when mutations disrupt this repair process or dysferlin is absent. While lipid binding by dysferlin's C2A domain (dysC2A) is considered fundamental to the membrane resealing process, the molecular mechanism of this interaction is not fully understood. By applying nonlinear surface-specific vibrational spectroscopy, we have successfully demonstrated that dysferlin's N-terminal C2A domain (dysC2A) alters its binding orientation in response to a membrane's lipid composition. These experiments reveal that dysC2A utilizes a generic electrostatic binding interaction to bind to most anionic lipid surfaces, inserting its calcium binding loops into the lipid surface while orienting its β-sheets 30-40° from surface normal. However, at lipid surfaces, where PI(4,5)P2 is present, dysC2A tilts its β-sheets more than 60° from surface normal to expose a polybasic face, while it binds to the PI(4,5)P2 surface. Both lipid binding mechanisms are shown to occur alongside dysC2A-induced lipid clustering. These different binding mechanisms suggest that dysC2A could provide a molecular cue to the larger dysferlin protein as to signal whether it is bound to the sarcolemma or another lipid surface.
Collapse
Affiliation(s)
- Andrew P Carpenter
- The School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, Oregon97331, United States
| | - Patricia Khuu
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon97331, United States
| | - Tobias Weidner
- Department of Chemistry, Aarhus University, 8000Aarhus C, Denmark
| | - Colin P Johnson
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis, Oregon97331, United States
| | - Steven J Roeters
- Department of Chemistry, Aarhus University, 8000Aarhus C, Denmark
| | - Joe E Baio
- The School of Chemical, Biological, and Environmental Engineering, Oregon State University, Corvallis, Oregon97331, United States
| |
Collapse
|
8
|
Anwar S, Yokota T. Morpholino-Mediated Exons 28-29 Skipping of Dysferlin and Characterization of Multiexon-skipped Dysferlin using RT-PCR, Immunoblotting, and Membrane Wounding Assay. Methods Mol Biol 2023; 2587:183-196. [PMID: 36401031 DOI: 10.1007/978-1-0716-2772-3_11] [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] [Indexed: 06/16/2023]
Abstract
Dysferlinopathies are a group of disabling muscular dystrophies that includes limb girdle muscular dystrophy type 2B (LGMD2B), Miyoshi myopathy, and distal myopathy with anterior tibial onset (DMAT) as the main phenotypes. They are associated with molecular defects in DYSF, which encodes dysferlin, a key player in sarcolemmal homeostasis. Previous investigations have suggested that exon skipping may be a promising therapy for many patients with dysferlinopathies. It was reported that exons 28-29 of DYSF are dispensable for dysferlin functions. Here, we present a method for multiexon skipping of DYSF exons 28-29 using a cocktail of two phosphorodiamidate morpholino oligomers (PMOs) on cells derived from a dystrophinopathy patient. Also, we describe assays to characterize the multiexon skipped dysferlin at several levels by using one-step RT-PCR, immunoblotting, and a membrane wounding assay.
Collapse
Affiliation(s)
- Saeed Anwar
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada.
- The Friends of Garrett Cumming Research and Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, AB, Canada.
| |
Collapse
|
9
|
Ivanova A, Smirnikhina S, Lavrov A. Dysferlinopathies: clinical and genetic variability. Clin Genet 2022; 102:465-473. [PMID: 36029111 DOI: 10.1111/cge.14216] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 08/12/2022] [Accepted: 08/18/2022] [Indexed: 11/30/2022]
Abstract
Dysferlinopathies are a clinically heterogeneous group of diseases caused by mutations in the DYSF gene encoding the dysferlin protein. Dysferlin is mostly expressed in muscle tissues and is localized in the sarcolemma, where it performs its main function of resealing and maintaining of the integrity of the cell membrane. At least four forms of dysferlinopathies have been described: Miyoshi myopathy, limb-girdle muscular dystrophy type 2B, distal myopathy with anterior tibial onset, and isolated hyperCKemia. Here we review the clinical features of different forms of dysferlinopathies and attempt to identify genotype-phenotype correlations. Because of the great clinical variability and rarety of the disease and mutations little is known, how different phenotypes develop as a result of different mutations. However missense mutations seem to induce more severe disease than LoF, which is typical for many muscle dystrophies. The role of several specific mutations and possible gene modifiers is also discussed in the paper.
Collapse
Affiliation(s)
- Alisa Ivanova
- Research Centre for Medical Genetics, Moskvorechye 1, Moscow, Russia
| | | | - Alexander Lavrov
- Research Centre for Medical Genetics, Moskvorechye 1, Moscow, Russia
| |
Collapse
|
10
|
Vahedi SM, Salek Ardestani S, Pahlevan Afshari K, Ghoreishifar SM, Moghaddaszadeh-Ahrabi S, Banabazi MH, Brito LF. Genome-Wide Selection Signatures and Human-Mediated Introgression Events in Bos taurus indicus-influenced Composite Beef Cattle. Front Genet 2022; 13:844653. [PMID: 35719394 PMCID: PMC9201998 DOI: 10.3389/fgene.2022.844653] [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: 12/28/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Genetic introgression from interbreeding hybridization of European Bos taurus taurus (EBT) and Indian Bos taurus indicus (IBI) cattle breeds have been widely used to combine the climatic resilience of the IBI cattle and the higher productivity of EBT when forming new composite beef cattle (CB) populations. The subsequent breeding strategies have shifted their initial genomic compositions. To uncover population structure, signatures of selection, and potential introgression events in CB populations, high-density genotypes [containing 492,954 single nucleotide polymorphisms (SNPs) after the quality control] of 486 individuals from 15 cattle breeds, including EBT, IBI, and CB populations, along with two Bos grunniens genotypes as outgroup were used in this study. Then, in-depth population genetics analyses were performed for three CB breeds of Beefmaster, Brangus, and Santa Gertrudis. Neighbor-joining, principal components, and admixture analyses confirmed the historical introgression of EBT and IBI haplotypes into CB breeds. The fdM statistics revealed that only 12.9% of CB populations' genetic components are of IBI origin. The results of signatures of selection analysis indicated different patterns of selection signals in the three CB breeds with primary pressure on pathways involved in protein processing and stress response in Beefmaster, cell proliferation regulation and immune response in Brangus, and amino acids and glucose metabolisms in Santa Gertrudis. An average of >90% of genomic regions underlying selection signatures were of EBT origin in the studied CB populations. Investigating the CB breeds' genome allows the estimation of EBT and IBI ancestral proportions and the locations within the genome where either taurine or indicine origin alleles are under selective pressure. Such findings highlight various opportunities to control the selection process more efficiently and explore complementarity at the genomic level in CB populations.
Collapse
Affiliation(s)
- Seyed Milad Vahedi
- Department of Animal Science and Aquaculture, Dalhousie University, Truro, NS, Canada
| | - Siavash Salek Ardestani
- Department of Animal Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Kian Pahlevan Afshari
- Department of Animal Sciences, Islamic Azad University, Varamin-Pishva Branch, Varamin, Iran
| | - Seyed Mohammad Ghoreishifar
- Department of Animal Science, University College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Sima Moghaddaszadeh-Ahrabi
- Department of Animal Science, Faculty of Agriculture and Natural Resources, Islamic Azad University, Tabriz Branch, Tabriz, Iran
| | - Mohammad Hossein Banabazi
- Department of Animal Breeding and Genetics (HGEN), Centre for Veterinary Medicine and Animal Science (VHC), Swedish University of Agricultural Sciences (SLU), Uppsala, Sweden
| | - Luiz Fernando Brito
- Department of Animal Sciences, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
11
|
Zhong H, Yu M, Lin P, Zhao Z, Zheng X, Xi J, Zhu W, Zheng Y, Zhang W, Lv H, Yan C, Hu J, Wang Z, Lu J, Zhao C, Luo S, Yuan Y. Molecular landscape of DYSF mutations in dysferlinopathy: From a Chinese multicenter analysis to a worldwide perspective. Hum Mutat 2021; 42:1615-1623. [PMID: 34559919 DOI: 10.1002/humu.24284] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 08/10/2021] [Accepted: 09/04/2021] [Indexed: 01/07/2023]
Abstract
Dysferlinopathy is one of the most common subgroup of autosomal recessive limb-girdle muscular dystrophies that is caused by mutations in DYSF gene. However, there is currently no worldwide comprehensive genetic analysis of DYSF variants. Through a national multicenter collaborative effort in China, we identified 222 DYSF variants with 40 novel variants from 245 patients. We then integrated DYSF variants from disease-related genetic databases including LOVD (n = 1020) and Clinvar (n = 1179), to depict the global landscape of disease-related DYSF variants. Normal-population-derived DSYF variants from gnomAD (n = 4318) and ChinaMAP (n = 13,330) were also analyzed in comparison. In Chinese patients, gender instead of genotype showed influence on the onset age of dysferlinopathy, with males showing an earlier age of onset. After integrative analysis, we identified two hotspot DYSF mutations, c.2997G>T in world patients and c.1375dup in Chinese patients, respectively. Both the pathogenic and likely pathogenic variants scattered on the whole gene length of DYSF. However, three specific domains (C2F-C2G-TM, DysF, and C2B-Ferl-C2C) contained variants at higher frequencies than reported in both the databases and Chinese patients. This study comprehensively collected available DYSF variant data, which may pave way for genetic counselling and future clinical trial design for gene therapies in dysferlinopathy.
Collapse
Affiliation(s)
- Huahua Zhong
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Meng Yu
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Pengfei Lin
- Department of Neurology, Shandong University Qilu Hospital, Jinan, Shandong Province, China
| | - Zhe Zhao
- Department of Neuromuscular Disorders, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xueying Zheng
- Department of Biostatistics, School of Public Health and Key Laboratory of Public Health Safety, Fudan University, Shanghai, China
| | - Jianying Xi
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Wenhua Zhu
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Yiming Zheng
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - He Lv
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Chuanzhu Yan
- Department of Neurology, Shandong University Qilu Hospital, Jinan, Shandong Province, China
| | - Jing Hu
- Department of Neuromuscular Disorders, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Zhaoxia Wang
- Department of Neurology, Peking University First Hospital, Beijing, China
| | - Jiahong Lu
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Chongbo Zhao
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Sushan Luo
- Department of Neurology, Huashan Hospital Fudan University, Shanghai, China.,National Center for Neurological Disorders, Shanghai, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing, China
| |
Collapse
|
12
|
Zhu F, Zhang F, Hu L, Liu H, Li Y. Integrated Genome and Transcriptome Sequencing to Solve a Neuromuscular Puzzle: Miyoshi Muscular Dystrophy and Early Onset Primary Dystonia in Siblings of the Same Family. Front Genet 2021; 12:672906. [PMID: 34276779 PMCID: PMC8283672 DOI: 10.3389/fgene.2021.672906] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/23/2021] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Neuromuscular disorders (NMD), many of which are hereditary, affect muscular function. Due to advances in high-throughput sequencing technologies, the diagnosis of hereditary NMDs has dramatically improved in recent years. METHODS AND RESULTS In this study, we report an family with two siblings exhibiting two different NMD, Miyoshi muscular dystrophy (MMD) and early onset primary dystonia (EOPD). Whole exome sequencing (WES) identified a novel monoallelic frameshift deletion mutation (dysferlin: c.4404delC/p.I1469Sfs∗17) in the Dysferlin gene in the index patient who suffered from MMD. This deletion was inherited from his unaffected father and was carried by his younger sister with EOPD. However, immunostaining staining revealed an absence of dysferlin expression in the proband's muscle tissue and thus suggested the presence of the second underlying mutant allele in dysferlin. Using integrated RNA sequencing (RNA-seq) and whole genome sequencing (WGS) of muscle tissue, a novel deep intronic mutation in dysferlin (dysferlin: c.5341-415A > G) was discovered in the index patient. This mutation caused aberrant mRNA splicing and inclusion of an additional pseudoexon (PE) which we termed PE48.1. This PE was inherited from his unaffected mother. PE48.1 inclusion altered the Dysferlin sequence, causing premature termination of translation. CONCLUSION Using integrated genome and transcriptome sequencing, we discovered hereditary MMD and EOPD affecting two siblings of same family. Our results added further weight to the combined use of RNA-seq and WGS as an important method for detection of deep intronic gene mutations, and suggest that integrated sequencing assays are an effective strategy for the diagnosis of hereditary NMDs.
Collapse
Affiliation(s)
- Feng Zhu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fengxiao Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lizhi Hu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haowen Liu
- Department of Neurology, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yahua Li
- Department of Respiratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
13
|
Annexins and Membrane Repair Dysfunctions in Muscular Dystrophies. Int J Mol Sci 2021; 22:ijms22105276. [PMID: 34067866 PMCID: PMC8155887 DOI: 10.3390/ijms22105276] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/11/2021] [Accepted: 05/12/2021] [Indexed: 11/16/2022] Open
Abstract
Muscular dystrophies constitute a group of genetic disorders that cause weakness and progressive loss of skeletal muscle mass. Among them, Miyoshi muscular dystrophy 1 (MMD1), limb girdle muscular dystrophy type R2 (LGMDR2/2B), and LGMDR12 (2L) are characterized by mutation in gene encoding key membrane-repair protein, which leads to severe dysfunctions in sarcolemma repair. Cell membrane disruption is a physiological event induced by mechanical stress, such as muscle contraction and stretching. Like many eukaryotic cells, muscle fibers possess a protein machinery ensuring fast resealing of damaged plasma membrane. Members of the annexins A (ANXA) family belong to this protein machinery. ANXA are small soluble proteins, twelve in number in humans, which share the property of binding to membranes exposing negatively-charged phospholipids in the presence of calcium (Ca2+). Many ANXA have been reported to participate in membrane repair of varied cell types and species, including human skeletal muscle cells in which they may play a collective role in protection and repair of the sarcolemma. Here, we discuss the participation of ANXA in membrane repair of healthy skeletal muscle cells and how dysregulation of ANXA expression may impact the clinical severity of muscular dystrophies.
Collapse
|
14
|
Retrospective analysis and reclassification of DYSF variants in a large French series of dysferlinopathy patients. Genet Med 2021; 23:1574-1577. [PMID: 33927379 DOI: 10.1038/s41436-021-01164-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 03/07/2021] [Accepted: 03/22/2021] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Recent evolution of sequencing technologies and the development of international standards in variant interpretation have profoundly changed the diagnostic approaches in clinical genetics. As a consequence, many variants that were initially claimed to be disease-causing can be now reclassified as benign or uncertain in light of the new data available. Unfortunately, the misclassified variants are still present in the scientific literature and variant databases, greatly interfering with interpretation of diagnostic sequencing results. Despite the urgent need, large-scale efforts to update the classifications of these variants are still not sufficient. METHODS We retrospectively analyzed 176 DYSF gene variants that were identified in dysferlinopathy patients referred to the Marseille Medical Genetics Department for diagnostic sequencing since 2001. RESULTS We reclassified all variants into five-tier American College of Medical Genetics and Genomics and the Association for Molecular Pathology (ACMG/AMP) pathogenicity classes, revealing changed pathogenicity for 17 variants. We then updated the information for the variants that have been previously published in the variant database and submitted 46 additional DYSF variants. CONCLUSION Besides direct benefit for dysferlinopathy diagnostics, our study contributes to the much needed effort to reanalyze variants from previously published cohorts and to work with curators of variant databases to update the entries for erroneously classified variants.
Collapse
|
15
|
Zhang H, Li Y, Cheng Q, Chen X, Yu Q, Li Z. Abnormal Expression of Dysferlin in Blood Monocytes Supports Primary Dysferlinopathy in Patients Confirmed by Genetic Analyses. Front Neurol 2021; 11:540098. [PMID: 33613410 PMCID: PMC7890250 DOI: 10.3389/fneur.2020.540098] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 10/28/2020] [Indexed: 12/11/2022] Open
Abstract
Objective: Dysferlin deficiency causes dysferlinopathy. This study aimed to expand the mutational spectrum of dysferlinopathies, to further study one case with diagnostic ambiguity, and to identify the diagnostic value of dysferlin expression in total peripheral blood mononuclear cells (PBMC). Methods: The clinical and molecular profiles of dysferlinopathies in eight Chinese patients were evaluated. We also conducted magnetic resonance imaging (6/8) and determined dysferlin protein expression in muscle (7/8) and PBMC (3/8). Results: Nine of the 13 DYSF mutations identified were novel. One patient was homozygous for the Gln111Ter mutation by genomic DNA sequencing but was found to be heterozygous by sequencing of cDNA from total PBMC. A daughter of this patient did not carry any Gln111Ter mutation. Abnormal muscle MRI with predominant involvement of the medial gastrocnemius and soleus muscle was observed in 5/6 patients. Dysferlin levels were significantly reduced (immunohistochemistry/immunoblot) or absent (immunohistochemistry) in muscle and total PBMC (26–39%) for most patients. Sarcoplasmic accumulation of dysferlin was detected in one patient. Conclusion: Genomic DNA sequencing detects frequent homozygous mutations, while fewer heterozygous mutations in cDNA are detected after posttranscription. Total PBMC may serve as an alternative to confirm diagnosis and to guide further testing in dysferlinopathies. Our results contribute to the mutational spectrum of dysferlinopathies.
Collapse
Affiliation(s)
- Huili Zhang
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Yaqin Li
- Department of Neurology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qiusheng Cheng
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xi Chen
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Qiuxia Yu
- Prenatal Diagnostic Center, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Ze Li
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
16
|
Li L, Jing Z, Cheng L, Liu W, Wang H, Xu Y, Zheng X, Yu X, Liu S. Compound heterozygous DYSF variants causing limb-girdle muscular dystrophy type 2B in a Chinese family. J Gene Med 2020; 22:e3272. [PMID: 32889728 DOI: 10.1002/jgm.3272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 08/22/2020] [Accepted: 08/29/2020] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND The dysferlin gene or the DYSF gene encodes the Ca2+ -dependent phospholipid-binding protein dysferlin, which belongs to the ferlin family and is associated with muscle membrane regeneration and repair. Variants in the DYSF gene are responsible for limb-girdle muscular dystrophy type 2B (LGMD2B), also called limb-girdle muscular dystrophy recessive 2 (LGMDR2), a rare subtype of muscular dystrophy involving progressive muscle weakness and atrophy. The present study aimed to identify the variants responsible for the clinical symptoms of a Chinese patient with limb girdle muscular dystrophies (LGMDs) and to explore the genotype-phenotype associations of LGMD2B. METHODS A series of clinical examinations, including blood tests, magnetic resonance imaging scans for the lower legs, electromyography and muscle biopsy, was performed on the proband diagnosed with muscular dystrophies. Whole exome sequencing was conducted to detect the causative variants, followed by Sanger sequencing to validate these variants. RESULTS We identified two compound heterozygous variants in the DYSF gene, c.1058 T>C, p.(Leu353Pro) in exon 12 and c.1461C>A/p.Cys487* in exon 16 in this proband, which were inherited from the father and mother, respectively. In silico analysis for these variants revealed deleterious results by PolyPhen-2 (Polymorphism Phenotyping v2; http://genetics.bwh.harvard.edu/pph2), SIFT (Sorting Intolerant From Tolerant; https://sift.bii.a-star.edu.sg), PROVEAN (Protein Variation Effect Analyzer; http://provean.jcvi.org/seq_submit.php) and MutationTaster (http://www.mutationtaster.org). In addition, the two compound heterozygous variants in the proband were absent in 100 control individuals who had an identical ethnic origin and were from the same region, suggesting that these variants may be the pathogenic variants responsible for the LGMD2B phenotypes for this proband. CONCLUSIONS The present study broadens our understanding of the mutational spectrum of the DYSF gene, which provides a deep insight into the pathogenesis of LGMDs and accelerates the development of a prenatal diagnosis.
Collapse
Affiliation(s)
- Liangshan Li
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China.,Department of Clinical Laboratory, Medical College of Qingdao University, Qingdao, China
| | - Zhongcui Jing
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Lei Cheng
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wenmiao Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Haiyan Wang
- Department of Blood Transfusion, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yinglei Xu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xueping Zheng
- Department of Neurology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaoling Yu
- Department of Pathophysiology, Medical College of Qingdao University, Qingdao, China
| | - Shiguo Liu
- Medical Genetic Department, The Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
17
|
Abstract
Ferlins are multiple-C2-domain proteins involved in Ca2+-triggered membrane dynamics within the secretory, endocytic and lysosomal pathways. In bony vertebrates there are six ferlin genes encoding, in humans, dysferlin, otoferlin, myoferlin, Fer1L5 and 6 and the long noncoding RNA Fer1L4. Mutations in DYSF (dysferlin) can cause a range of muscle diseases with various clinical manifestations collectively known as dysferlinopathies, including limb-girdle muscular dystrophy type 2B (LGMD2B) and Miyoshi myopathy. A mutation in MYOF (myoferlin) was linked to a muscular dystrophy accompanied by cardiomyopathy. Mutations in OTOF (otoferlin) can be the cause of nonsyndromic deafness DFNB9. Dysregulated expression of any human ferlin may be associated with development of cancer. This review provides a detailed description of functions of the vertebrate ferlins with a focus on muscle ferlins and discusses the mechanisms leading to disease development.
Collapse
|
18
|
Verwey N, Gazzoli I, Krause S, Mamchaoui K, Mouly V, Aartsma-Rus A. Antisense-Mediated Skipping of Dysferlin Exons in Control and Dysferlinopathy Patient-Derived Cells. Nucleic Acid Ther 2019; 30:71-79. [PMID: 31873062 DOI: 10.1089/nat.2019.0788] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Dysferlinopathies encompass a spectrum of progressive muscular dystrophies caused by the lack of dysferlin due to missense mutations in the dysferlin gene or mutations causing premature truncation of protein translation. Dysferlin is a modular protein, and dysferlins lacking one or more repetitive domains have been shown to retain functionality. As such, antisense-mediated exon skipping has been proposed as a therapy for dysferlinopathy. By skipping the mutated exon, the reading frame would be maintained, while the mutation would be bypassed, thus allowing production of an internally deleted, but partially functional, dysferlin. We previously showed that dysferlin exon skipping is feasible in control cell lines. We here evaluated exon skipping and dysferlin protein restoration in patient-derived cells requiring the skipping of exon 9, 29, 30, or 34. Exon 30 skipping was possible at high efficiency, but did not result in increased dysferlin. We discovered that the alleged exon 30 mutation was in fact a polymorphism and identified a splicing mutation in intron 28 as the disease-causing mutation. While exon skipping was feasible for each of the other cell lines, no increases in dysferlin protein could be detected by western blotting.
Collapse
Affiliation(s)
- Nisha Verwey
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Isabella Gazzoli
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Sabine Krause
- Department of Neurology, Friedrich-Baur-Institute, Ludwig-Maximilians-University of Munich, Munchen, Germany
| | - Kamel Mamchaoui
- Sorbonne Université, INSERM, Institut de Myologie, Myology Research Center, CRM, Paris, France
| | - Vincent Mouly
- Sorbonne Université, INSERM, Institut de Myologie, Myology Research Center, CRM, Paris, France
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
19
|
Dominov JA, Uyan Ö, McKenna‐Yasek D, Nallamilli BRR, Kergourlay V, Bartoli M, Levy N, Hudson J, Evangelista T, Lochmuller H, Krahn M, Rufibach L, Hegde M, Brown RH. Correction of pseudoexon splicing caused by a novel intronic dysferlin mutation. Ann Clin Transl Neurol 2019; 6:642-654. [PMID: 31019989 PMCID: PMC6469257 DOI: 10.1002/acn3.738] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2018] [Revised: 01/12/2019] [Accepted: 01/21/2019] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE Dysferlin is a large transmembrane protein that functions in critical processes of membrane repair and vesicle fusion. Dysferlin-deficiency due to mutations in the dysferlin gene leads to muscular dystrophy (Miyoshi myopathy (MM), limb girdle muscular dystrophy type 2B (LGMD2B), distal myopathy with anterior tibial onset (DMAT)), typically with early adult onset. At least 416 pathogenic dysferlin mutations are known, but for approximately 17% of patients, one or both of their pathogenic variants remain undefined following standard exon sequencing methods that interrogate exons and nearby flanking intronic regions but not the majority of intronic regions. METHODS We sequenced RNA from myogenic cells to identify a novel dysferlin pathogenic variant in two affected siblings that previously had only one disease-causing variant identified. We designed antisense oligonucleotides (AONs) to bypass the effects of this mutation on RNA splicing. RESULTS We identified a new pathogenic point mutation deep within dysferlin intron 50i. This intronic variant causes aberrant mRNA splicing and inclusion of an additional pseudoexon (PE, we term PE50.1) within the mature dysferlin mRNA. PE50.1 inclusion alters the protein sequence, causing premature translation termination. We identified this mutation in 23 dysferlinopathy patients (seventeen families), revealing it to be one of the more prevalent dysferlin mutations. We used AON-mediated exon skipping to correct the aberrant PE50.1 splicing events in vitro, which increased normal mRNA production and significantly restored dysferlin protein expression. INTERPRETATION Deep intronic mutations can be a common underlying cause of dysferlinopathy, and importantly, could be treatable with AON-based exon-skipping strategies.
Collapse
Affiliation(s)
- Janice A. Dominov
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts
| | - Özgün Uyan
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts
| | - Diane McKenna‐Yasek
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts
| | - Babi Ramesh Reddy Nallamilli
- Department of Human GeneticsEmory University School of MedicineAtlantaGeorgia
- Present address:
Perkin Elmer GenomicsWalthamMassachusetts
| | - Virginie Kergourlay
- Marseille Medical Genetics ‐ Translational NeuromyologyAix‐Marseille UnivINSERMMMGMarseilleFrance
| | - Marc Bartoli
- Marseille Medical Genetics ‐ Translational NeuromyologyAix‐Marseille UnivINSERMMMGMarseilleFrance
| | - Nicolas Levy
- Marseille Medical Genetics ‐ Translational NeuromyologyAix‐Marseille UnivINSERMMMGMarseilleFrance
- Département de Génétique MédicaleAPHMHôpital Timone EnfantsMarseilleFrance
| | - Judith Hudson
- Northern Molecular Genetics ServiceNewcastle upon TyneUnited Kingdom
| | - Teresinha Evangelista
- Newcastle University John Walton Centre for Muscular Dystrophy ResearchMRC Centre for Neuromuscular DiseasesInstitute of Genetic MedicineNewcastle upon TyneUnited Kingdom
| | - Hanns Lochmuller
- Newcastle University John Walton Centre for Muscular Dystrophy ResearchMRC Centre for Neuromuscular DiseasesInstitute of Genetic MedicineNewcastle upon TyneUnited Kingdom
- Department of Neuropediatrics and Muscle DisordersFaculty of MedicineMedical Center–University of FreiburgFreiburgGermany
- Centro Nacional de Análisis Genómico (CNAG‐CRG)Center for Genomic RegulationBarcelona Institute of Science and Technology (BIST)BarcelonaCataloniaSpain
- Children's Hospital of Eastern Ontario Research InstituteUniversity of OttawaOttawaCanada
- Division of NeurologyDepartment of MedicineThe Ottawa HospitalOttawaCanada
| | - Martin Krahn
- Marseille Medical Genetics ‐ Translational NeuromyologyAix‐Marseille UnivINSERMMMGMarseilleFrance
- Département de Génétique MédicaleAPHMHôpital Timone EnfantsMarseilleFrance
| | | | - Madhuri Hegde
- Department of Human GeneticsEmory University School of MedicineAtlantaGeorgia
| | - Robert H. Brown
- Department of NeurologyUniversity of Massachusetts Medical SchoolWorcesterMassachusetts
| |
Collapse
|
20
|
Yauy K, Baux D, Pegeot H, Van Goethem C, Mathieu C, Guignard T, Juntas Morales R, Lacourt D, Krahn M, Lehtokari VL, Bonne G, Tuffery-Giraud S, Koenig M, Cossée M. MoBiDiC Prioritization Algorithm, a Free, Accessible, and Efficient Pipeline for Single-Nucleotide Variant Annotation and Prioritization for Next-Generation Sequencing Routine Molecular Diagnosis. J Mol Diagn 2018; 20:465-473. [PMID: 29689380 DOI: 10.1016/j.jmoldx.2018.03.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 02/16/2018] [Accepted: 03/09/2018] [Indexed: 12/22/2022] Open
Abstract
Interpretation of next-generation sequencing constitutes the main limitation of molecular diagnostics. In diagnosing myopathies and muscular dystrophies, another issue is efficiency in predicting the pathogenicity of variants identified in large genes, especially TTN; current in silico prediction tools show limitations in predicting and ranking the numerous variants of such genes. We propose a variant-prioritization tool, the MoBiDiCprioritization algorithm (MPA). MPA is based on curated interpretation of data on previously reported variants, biological assumptions, and splice and missense predictors, and is used to prioritize all types of single-nucleotide variants. MPA was validated by comparing its sensitivity and specificity to those of dbNSFP database prediction tools, using a data set composed of DYSF, DMD, LMNA, NEB, and TTN variants extracted from expert-reviewed and ExAC databases. MPA obtained the best annotation rates for missense and splice variants. As MPA aggregates the results from several predictors, individual predictor errors are counterweighted, improving the sensitivity and specificity of missense and splice variant predictions. We propose a sequential use of MPA, beginning with the selection of variants with higher scores and followed by, in the absence of candidate pathologic variants, consideration of variants with lower scores. We provide scripts and documentation for free academic use and a validated annotation pipeline scaled for panel and exome sequencing to prioritize single-nucleotide variants from a VCF file.
Collapse
Affiliation(s)
- Kevin Yauy
- Laboratoire de Génétique Moléculaire, CHU Montpellier, Montpellier, France.
| | - David Baux
- Laboratoire de Génétique Moléculaire, CHU Montpellier, Montpellier, France
| | - Henri Pegeot
- Laboratoire de Génétique Moléculaire, CHU Montpellier, Montpellier, France
| | - Charles Van Goethem
- Laboratoire de Biopathologie Cellulaire et Tissulaire des Tumeurs, Hôpital Arnaud de Villeneuve, CHU Montpellier, Montpellier, France
| | - Charly Mathieu
- Laboratoire de Génétique Moléculaire, CHU Montpellier, Montpellier, France
| | - Thomas Guignard
- Plateforme Recherche de Microremaniements Chromosomiques-Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, CHU Montpellier, Montpellier, France; Faculté de Médecine Montpellier-Nîmes, Université de Montpellier, Montpellier, France
| | | | - Delphine Lacourt
- Laboratoire de Génétique Moléculaire, CHU Montpellier, Montpellier, France
| | - Martin Krahn
- Unité de Génétique Médicale et Génomique Fonctionnelle INSERM UMRS910, Université d'Aix Marseille, Marseille, France; Département de Génétique Médicale, Hôpital Timone Enfants, Assistance Publique Hôpitaux de Marseille, Marseille, France
| | - Vilma-Lotta Lehtokari
- The Folkhalsan Institute of Genetics and the Department of Medical Genetics, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - Gisele Bonne
- Unité INSERM U974-Thérapie des Maladies du Muscle Striée, Center of Research in Myology, Institut de Myologie, Université Pierre et Marie Curie, Sorbonne Universités, Paris, France
| | - Sylvie Tuffery-Giraud
- Laboratoire de Génétique des Maladies Rares EA7402, Université de Montpellier, Montpellier, France
| | - Michel Koenig
- Laboratoire de Génétique Moléculaire, CHU Montpellier, Montpellier, France; Laboratoire de Génétique des Maladies Rares EA7402, Université de Montpellier, Montpellier, France
| | - Mireille Cossée
- Laboratoire de Génétique Moléculaire, CHU Montpellier, Montpellier, France; Laboratoire de Génétique des Maladies Rares EA7402, Université de Montpellier, Montpellier, France
| |
Collapse
|
21
|
Dastur RS, Gaitonde PS, Kachwala M, Nallamilli BRR, Ankala A, Khadilkar SV, Atchayaram N, Gayathri N, Meena AK, Rufibach L, Shira S, Hegde M. Detection of Dysferlin Gene Pathogenic Variants in the Indian Population in Patients Predicted to have a Dysferlinopathy Using a Blood-based Monocyte Assay and Clinical Algorithm: A Model for Accurate and Cost-effective Diagnosis. Ann Indian Acad Neurol 2017; 20:302-308. [PMID: 28904466 PMCID: PMC5586129 DOI: 10.4103/aian.aian_129_17] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Limb-girdle muscular dystrophy (LGMD) is the most common adult-onset class of muscular dystrophies in India, but a majority of suspected LGMDs in India remain unclassified to the genetic subtype level. The next-generation sequencing (NGS)-based approaches have allowed molecular characterization and subtype diagnosis in a majority of these patients in India. MATERIALS AND METHODS (I) To select probable dysferlinopathy (LGMD2B) cases from other LGMD subtypes using two screening methods (i) to determine the status of dysferlin protein expression in blood (peripheral blood mononuclear cell) by monocyte assay (ii) using a predictive algorithm called automated LGMD diagnostic assistant (ALDA) to obtain possible LGMD subtypes based on clinical symptoms. (II) Identification of gene pathogenic variants by NGS for 34 genes associated with LGMD or LGMD like muscular dystrophies, in cases showing: absence of dysferlin protein by the monocyte assay and/or a typical dysferlinopathy phenotype, with medium to high predictive scores using the ALDA tool. RESULTS Out of the 125 patients screened by NGS, 96 were confirmed with two dysferlin variants, of which 84 were homozygous. Single dysferlin pathogenic variants were seen in 4 patients, whereas 25 showed no variants in the dysferlin gene. CONCLUSION In this study, 98.2% of patients with absence of the dysferlin protein showed one or more variants in the dysferlin gene and hence has a high predictive significance in diagnosing dysferlinopathies. However, collection of blood samples from all over India for protein analysis is expensive. Our analysis shows that the use of the "ALDA tool" could be a cost-effective alternative method. Identification of dysferlin pathogenic variants by NGS is the ultimate method for diagnosing dysferlinopathies though follow-up with the monocyte assay can be useful to understand the phenotype in relation to the dysferlin protein expression and also be a useful biomarker for future clinical trials.
Collapse
Affiliation(s)
- Rashna Sam Dastur
- Centre for Advanced Molecular Diagnostics in Neuromuscular Disorders, Atlanta, Georgia, USA
| | | | - Munira Kachwala
- Centre for Advanced Molecular Diagnostics in Neuromuscular Disorders, Atlanta, Georgia, USA
| | - Babi R. R. Nallamilli
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Arunkanth Ankala
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Satish V. Khadilkar
- Department of Neurology, Sir J J Group of Hospitals, Grant Medical College, Mumbai, Maharashtra, India
| | | | - N. Gayathri
- Department of Neurology, NIMHANS, Bengaluru, Karnataka
| | - A. K. Meena
- Department of Neurology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | | | | | - Madhuri Hegde
- Department of Human Genetics, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
22
|
Fanin M, Angelini C. Progress and challenges in diagnosis of dysferlinopathy. Muscle Nerve 2016; 54:821-835. [DOI: 10.1002/mus.25367] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2016] [Indexed: 01/22/2023]
Affiliation(s)
- Marina Fanin
- Department of Neurosciences; University of Padova; Biomedical Campus “Pietro d'Abano”, via Giuseppe Orus 2B 35129 Padova Italy
| | | |
Collapse
|
23
|
Middel V, Zhou L, Takamiya M, Beil T, Shahid M, Roostalu U, Grabher C, Rastegar S, Reischl M, Nienhaus GU, Strähle U. Dysferlin-mediated phosphatidylserine sorting engages macrophages in sarcolemma repair. Nat Commun 2016; 7:12875. [PMID: 27641898 PMCID: PMC5031802 DOI: 10.1038/ncomms12875] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 08/10/2016] [Indexed: 01/22/2023] Open
Abstract
Failure to repair the sarcolemma leads to muscle cell death, depletion of stem cells and myopathy. Hence, membrane lesions are instantly sealed by a repair patch consisting of lipids and proteins. It has remained elusive how this patch is removed to restore cell membrane integrity. Here we examine sarcolemmal repair in live zebrafish embryos by real-time imaging. Macrophages remove the patch. Phosphatidylserine (PS), an 'eat-me' signal for macrophages, is rapidly sorted from adjacent sarcolemma to the repair patch in a Dysferlin (Dysf) dependent process in zebrafish and human cells. A previously unrecognized arginine-rich motif in Dysf is crucial for PS accumulation. It carries mutations in patients presenting with limb-girdle muscular dystrophy 2B. This underscores the relevance of this sequence and uncovers a novel pathophysiological mechanism underlying this class of myopathies. Our data show that membrane repair is a multi-tiered process involving immediate, cell-intrinsic mechanisms as well as myofiber/macrophage interactions.
Collapse
Affiliation(s)
- Volker Middel
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Lu Zhou
- Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), Wolfgang-Gaede-Straße 1, 76131 Karlsruhe, Germany.,Institute of Nanotechnology, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Masanari Takamiya
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Tanja Beil
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Maryam Shahid
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Urmas Roostalu
- Institute of Inflammation and Repair, The University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Clemens Grabher
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Sepand Rastegar
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Markus Reischl
- Institute for Applied Computer Science, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| | - Gerd Ulrich Nienhaus
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany.,Institute of Applied Physics, Karlsruhe Institute of Technology (KIT), Wolfgang-Gaede-Straße 1, 76131 Karlsruhe, Germany.,Institute of Nanotechnology, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany.,Department of Physics, University of Illinois at Urbana-Champaign, 1110 West Green Street, 61801 Urbana, Illinois, US
| | - Uwe Strähle
- Institute of Toxicology and Genetics, Karlsruhe Institute of Technology (KIT), PO Box 3640, 76021 Karlsruhe, Germany
| |
Collapse
|
24
|
Kergourlay V, Blandin G, Blanck V, Lévy N, Bartoli M, Krahn M. Comment on: A novel dysferlin-mutant pseudoexon bypassed with antisense oligonucleotides. Ann Clin Transl Neurol 2015; 2:783-4. [PMID: 26273692 PMCID: PMC4531062 DOI: 10.1002/acn3.216] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Virginie Kergourlay
- Aix Marseille Université, GMGF 13385, Marseille, France ; Inserm, UMR_S 910 13385, Marseille, France
| | - Gaëlle Blandin
- Aix Marseille Université, GMGF 13385, Marseille, France ; Inserm, UMR_S 910 13385, Marseille, France
| | - Véronique Blanck
- AP-HM, Département de Génétique Médicale et de Biologie Cellulaire, Hôpital d'Enfants de la Timone 13385, Marseille, France
| | - Nicolas Lévy
- Aix Marseille Université, GMGF 13385, Marseille, France ; Inserm, UMR_S 910 13385, Marseille, France ; AP-HM, Département de Génétique Médicale et de Biologie Cellulaire, Hôpital d'Enfants de la Timone 13385, Marseille, France
| | - Marc Bartoli
- Aix Marseille Université, GMGF 13385, Marseille, France ; Inserm, UMR_S 910 13385, Marseille, France ; AP-HM, Département de Génétique Médicale et de Biologie Cellulaire, Hôpital d'Enfants de la Timone 13385, Marseille, France
| | - Martin Krahn
- Aix Marseille Université, GMGF 13385, Marseille, France ; Inserm, UMR_S 910 13385, Marseille, France ; AP-HM, Département de Génétique Médicale et de Biologie Cellulaire, Hôpital d'Enfants de la Timone 13385, Marseille, France
| |
Collapse
|
25
|
Lazar CH, Kimchi A, Namburi P, Mutsuddi M, Zelinger L, Beryozkin A, Ben-Simhon S, Obolensky A, Ben-Neriah Z, Argov Z, Pikarsky E, Fellig Y, Marks-Ohana D, Ratnapriya R, Banin E, Sharon D, Swaroop A. Nonsyndromic Early-Onset Cone-Rod Dystrophy and Limb-Girdle Muscular Dystrophy in a Consanguineous Israeli Family are Caused by Two Independent yet Linked Mutations in ALMS1 and DYSF. Hum Mutat 2015; 36:836-41. [PMID: 26077327 DOI: 10.1002/humu.22822] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Accepted: 06/09/2015] [Indexed: 01/26/2023]
Abstract
Genetic analysis of clinical phenotypes in consanguineous families is complicated by coinheritance of large DNA regions carrying independent variants. Here, we characterized a family with early onset cone-rod dystrophy (CRD) and muscular dystrophy. Homozygosity mapping (HM) followed by whole exome sequencing revealed a nonsense mutation, p.R270*, in ALMS1 and two novel potentially disease-causing missense variants, p.R1581C and p.Y2070C, in DYSF. ALMS1 and DYSF are genetically and physically linked on chromosome 2 in a genomic region suggested by HM and associated with Alström syndrome, which includes CRD, and with limb girdle muscular dystrophy, respectively. Affected family members lack additional systemic manifestations of Alström syndrome but exhibit mild muscular dystrophy. RNA-seq data did not reveal any significant variations in ALMS1 transcripts in the human retina. Our study thus implicates ALMS1 as a nonsyndromic retinal disease gene and suggests a potential role of variants in interacting cilia genes in modifying clinical phenotypes.
Collapse
Affiliation(s)
- Csilla H Lazar
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland.,Molecular Biology Center, Interdisciplinary Research Institute on Bio-Nano Sciences, Babes-Bolyai-University, Cluj-Napoca, Romania
| | - Adva Kimchi
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Prasanthi Namburi
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Mousumi Mutsuddi
- Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi, India
| | - Lina Zelinger
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland.,Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Avigail Beryozkin
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Shiran Ben-Simhon
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Alexey Obolensky
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ziva Ben-Neriah
- Department of Genetics, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Zohar Argov
- Department of Neurology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Eli Pikarsky
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Yakov Fellig
- Department of Pathology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Devorah Marks-Ohana
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Rinki Ratnapriya
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| | - Eyal Banin
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Dror Sharon
- Department of Ophthalmology, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Anand Swaroop
- Neurobiology-Neurodegeneration & Repair Laboratory, National Eye Institute, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
26
|
Woudt L, Di Capua GA, Krahn M, Castiglioni C, Hughes R, Campero M, Trangulao A, González-Hormazábal P, Godoy-Herrera R, Lévy N, Urtizberea JA, Jara L, Bevilacqua JA. Toward an objective measure of functional disability in dysferlinopathy. Muscle Nerve 2015; 53:49-57. [DOI: 10.1002/mus.24685] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2015] [Indexed: 11/06/2022]
Affiliation(s)
- Lisanne Woudt
- Unidad Neuromuscular; Departamento de Neurología y Neurocirugía; Hospital Clínico Universidad de Chile, Santos Dumont 999, 2do. piso; Sector E. Independencia 8380456 Santiago Chile
| | - Gabriella A. Di Capua
- Programa de Genética Humana; Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile; Santiago Chile
| | - Martin Krahn
- Aix Marseille Université; INSERM, Medical Genetics and Functional Genomics; Unité Mixte de Recherche_S 910 Marseille France
- Assistance Publique - Hôpitaux de Marseille; Département de Génétique Médicale; Hôpital Timone Enfants; Marseille France
| | - Claudia Castiglioni
- Unidad de Neurología; Departamento de Pediatría; Clínica Las Condes; Santiago Chile
| | - Ricardo Hughes
- Unidad Neuromuscular; Departamento de Neurología y Neurocirugía; Hospital Clínico Universidad de Chile, Santos Dumont 999, 2do. piso; Sector E. Independencia 8380456 Santiago Chile
| | - Mario Campero
- Unidad Neuromuscular; Departamento de Neurología y Neurocirugía; Hospital Clínico Universidad de Chile, Santos Dumont 999, 2do. piso; Sector E. Independencia 8380456 Santiago Chile
| | - Alejandra Trangulao
- Programa Anatomía y Biología del Desarrollo; Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile; Santiago Chile
| | - Patricio González-Hormazábal
- Programa de Genética Humana; Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile; Santiago Chile
| | - Raúl Godoy-Herrera
- Programa de Genética Humana; Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile; Santiago Chile
| | - Nicolas Lévy
- Aix Marseille Université; INSERM, Medical Genetics and Functional Genomics; Unité Mixte de Recherche_S 910 Marseille France
- Assistance Publique - Hôpitaux de Marseille; Département de Génétique Médicale; Hôpital Timone Enfants; Marseille France
| | - Jon Andoni Urtizberea
- Unité Neuromusculaire, Hôpital Marin de Hendaye; Assistance Publique - Hôpitaux de Paris Hendaye France
| | - Lilian Jara
- Programa de Genética Humana; Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile; Santiago Chile
| | - Jorge A. Bevilacqua
- Unidad Neuromuscular; Departamento de Neurología y Neurocirugía; Hospital Clínico Universidad de Chile, Santos Dumont 999, 2do. piso; Sector E. Independencia 8380456 Santiago Chile
- Programa Anatomía y Biología del Desarrollo; Instituto de Ciencias Biomédicas (ICBM), Facultad de Medicina, Universidad de Chile; Santiago Chile
| |
Collapse
|
27
|
Kergourlay V, Raï G, Blandin G, Salgado D, Béroud C, Lévy N, Krahn M, Bartoli M. Identification of Splicing Defects Caused by Mutations in the Dysferlin Gene. Hum Mutat 2014; 35:1532-41. [DOI: 10.1002/humu.22710] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 10/03/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Virginie Kergourlay
- Aix Marseille Université; GMGF; Marseille 13385 France
- Inserm, UMR_S 910; Marseille 13385 France
| | - Ghadi Raï
- Aix Marseille Université; GMGF; Marseille 13385 France
- Inserm, UMR_S 910; Marseille 13385 France
| | - Gaëlle Blandin
- Aix Marseille Université; GMGF; Marseille 13385 France
- Inserm, UMR_S 910; Marseille 13385 France
| | - David Salgado
- Aix Marseille Université; GMGF; Marseille 13385 France
- Inserm, UMR_S 910; Marseille 13385 France
| | - Christophe Béroud
- Aix Marseille Université; GMGF; Marseille 13385 France
- Inserm, UMR_S 910; Marseille 13385 France
- Département de Génétique Médicale et de Biologie Cellulaire; AP-HM, Hôpital d'Enfants de la Timone; Marseille 13385 France
| | - Nicolas Lévy
- Aix Marseille Université; GMGF; Marseille 13385 France
- Inserm, UMR_S 910; Marseille 13385 France
- Département de Génétique Médicale et de Biologie Cellulaire; AP-HM, Hôpital d'Enfants de la Timone; Marseille 13385 France
| | - Martin Krahn
- Aix Marseille Université; GMGF; Marseille 13385 France
- Inserm, UMR_S 910; Marseille 13385 France
- Département de Génétique Médicale et de Biologie Cellulaire; AP-HM, Hôpital d'Enfants de la Timone; Marseille 13385 France
| | - Marc Bartoli
- Aix Marseille Université; GMGF; Marseille 13385 France
- Inserm, UMR_S 910; Marseille 13385 France
- Département de Génétique Médicale et de Biologie Cellulaire; AP-HM, Hôpital d'Enfants de la Timone; Marseille 13385 France
| |
Collapse
|
28
|
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: 2.1] [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
|
29
|
Gallardo E, Ankala A, Núñez-Álvarez Y, Hegde M, Diaz-Manera J, Luna ND, Pastoret A, Suelves M, Illa I. Genetic and Epigenetic Determinants of Low Dysferlin Expression in Monocytes. Hum Mutat 2014; 35:990-7. [DOI: 10.1002/humu.22591] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 05/02/2014] [Indexed: 01/23/2023]
Affiliation(s)
- Eduard Gallardo
- Laboratori de Malalties Neuromusculars; Institut de Recerca de HSCSP; Universitat Autònoma de Barcelona (UAB); Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
| | - Arunkanth Ankala
- Department of Human Genetics; Emory University School of Medicine; Atlanta Georgia
| | - Yaiza Núñez-Álvarez
- Institut de Medicina Predictiva i Personalitzada del Càncer (IMPPC) i Institut Germans Trias i Pujol (IGTP); Badalona Spain
| | - Madhuri Hegde
- Department of Human Genetics; Emory University School of Medicine; Atlanta Georgia
| | - Jordi Diaz-Manera
- Laboratori de Malalties Neuromusculars; Institut de Recerca de HSCSP; Universitat Autònoma de Barcelona (UAB); Barcelona Spain
- Servei de Neurologia; Hospital de Sant Pau; Universitat Autònoma de Barcelona (UAB); Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
| | - Noemí De Luna
- Laboratori de Patologia Mitocondrial i Neuromuscular; Hospital Universitari Vall d'Hebron, Institut de Recerca (VHIR); Universitat Autònoma de Barcelona
- Centre for Biomedical Network Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III; Valencia Spain
| | - Ana Pastoret
- Laboratori de Malalties Neuromusculars; Institut de Recerca de HSCSP; Universitat Autònoma de Barcelona (UAB); Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
| | - Mònica Suelves
- Institut de Medicina Predictiva i Personalitzada del Càncer (IMPPC) i Institut Germans Trias i Pujol (IGTP); Badalona Spain
| | - Isabel Illa
- Laboratori de Malalties Neuromusculars; Institut de Recerca de HSCSP; Universitat Autònoma de Barcelona (UAB); Barcelona Spain
- Servei de Neurologia; Hospital de Sant Pau; Universitat Autònoma de Barcelona (UAB); Barcelona Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED); Madrid Spain
| |
Collapse
|
30
|
Touznik A, Lee JJA, Yokota T. New developments in exon skipping and splice modulation therapies for neuromuscular diseases. Expert Opin Biol Ther 2014; 14:809-19. [PMID: 24620745 DOI: 10.1517/14712598.2014.896335] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Antisense oligonucleotide (AON) therapy is a form of treatment for genetic or infectious diseases using small, synthetic DNA-like molecules called AONs. Recent advances in the development of AONs that show improved stability and increased sequence specificity have led to clinical trials for several neuromuscular diseases. Impressive preclinical and clinical data are published regarding the usage of AONs in exon-skipping and splice modulation strategies to increase dystrophin production in Duchenne muscular dystrophy (DMD) and survival of motor neuron (SMN) production in spinal muscular atrophy (SMA). AREAS COVERED In this review, we focus on the current progress and challenges of exon-skipping and splice modulation therapies. In addition, we discuss the recent failure of the Phase III clinical trials of exon 51 skipping (drisapersen) for DMD. EXPERT OPINION The main approach of AON therapy in DMD and SMA is to rescue ('knock up' or increase) target proteins through exon skipping or exon inclusion; conversely, most conventional antisense drugs are designed to knock down (inhibit) the target. Encouraging preclinical data using this 'knock up' approach are also reported to rescue dysferlinopathies, including limb-girdle muscular dystrophy type 2B, Miyoshi myopathy, distal myopathy with anterior tibial onset and Fukuyama congenital muscular dystrophy.
Collapse
Affiliation(s)
- Aleksander Touznik
- University of Alberta, Faculty of Medicine and Dentistry, Department of Medical Genetics , Edmonton, Alberta , Canada
| | | | | |
Collapse
|
31
|
Joshi PR, Gläser D, Dreßel C, Kress W, Weis J, Deschauer M. Anoctamin 5 muscular dystrophy associated with a silent p.Leu115Leu mutation resulting in exon skipping. Neuromuscul Disord 2013; 24:43-7. [PMID: 24239059 DOI: 10.1016/j.nmd.2013.09.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2013] [Revised: 07/22/2013] [Accepted: 09/02/2013] [Indexed: 10/26/2022]
Abstract
We report a 45year-old patient with an asymmetrical proximal muscle weakness affecting the quadriceps muscle of the right leg starting at the age of 32years. CK was 25-fold increased. MRI of the legs showed signs of fatty degeneration more pronounced in the right side. Biopsy of a thigh muscle showed dystrophic pattern and amyloid deposition in blood vessel walls. The coding region and exon/intron boundaries of the ANO5 gene were amplified and sequenced. The common c.191dupA mutation and a silent novel p.Leu115Leu (c.345G>A) variant were identified. This silent variant was listed neither in the LOVD database nor in the SNP database. To evaluate the pathogenicity of the novel silent mutation in ANO5, cDNA analysis was performed that demonstrated skipping of exon 6. So far, no case with a silent mutation leading to abnormal splicing has been identified in Anoctamin 5 muscular dystrophy. Present findings emphasize that cDNA analysis should be done if a silent variant is not annotated in the databases. In Anoctamin 5 muscular dystrophy a molecular diagnosis is even more important as protein investigation through Western blotting or immunohistochemistry is not yet established.
Collapse
Affiliation(s)
- Pushpa Raj Joshi
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Germany.
| | | | | | - Wolfram Kress
- Institute of Human Genetics, Biozentrum, University Würzburg, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen, Germany
| | - Marcus Deschauer
- Department of Neurology, Martin-Luther-University Halle-Wittenberg, Germany
| |
Collapse
|
32
|
Mariano A, Henning A, Han R. Dysferlin-deficient muscular dystrophy and innate immune activation. FEBS J 2013; 280:4165-76. [PMID: 23527661 DOI: 10.1111/febs.12261] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 03/06/2013] [Accepted: 03/20/2013] [Indexed: 11/27/2022]
Abstract
Cells encounter many physical, chemical and biological stresses that perturb plasma membrane integrity, warranting an immediate membrane repair response to regain cell homeostasis. Failure to respond properly to such perturbation leads to individual cell death, which may also produce systemic influence by triggering sterile immunological responses. In this review, we discuss recent progress on understanding the mechanisms underlying muscle cell membrane repair and the potential mediators of innate immune activation when the membrane repair system is defective, specifically focusing on pathology associated with dysferlin deficiency.
Collapse
Affiliation(s)
- Andrew Mariano
- Department of Cell and Molecular Physiology, Loyola University Chicago Health Science Division, Maywood, IL 60153, USA
| | | | | |
Collapse
|
33
|
Zhao Z, Hu J, Sakiyama Y, Okamoto Y, Higuchi I, Li N, Shen H, Takashima H. DYSF mutation analysis in a group of Chinese patients with dysferlinopathy. Clin Neurol Neurosurg 2012; 115:1234-7. [PMID: 23254335 DOI: 10.1016/j.clineuro.2012.11.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2011] [Revised: 11/11/2012] [Accepted: 11/18/2012] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Dysferlinopathies belong to heterogeneous group of autosomal recessive muscular disorders caused by mutations in the gene encoding dysferlin. The classifications of the dysferlinopathies mainly include limb-girdle muscular dystrophy 2B (LGMD2B) with predominantly proximal weakness, Miyoshi myopathy (MM) with calf muscle weakness and atrophy, and distal myopathy with anterior tibial onset (DMAT) with tibialis muscle atrophy. We describe the genetic character of dysferlinopathies in a group of Chinese patients. METHODS DYSF mutations screening were done after muscle biopsy and immunohistochemical staining. RESULTS Eight patients showed an absence or drastic decrease of dysferlin expression in biopsied muscle. We identified 6 different mutations, including one nonsense mutation, two insertion mutation, two deletion mutations and one splice site mutation. Five of them were novel mutations. CONCLUSION We described 8 Chinese patients with dysferlinopathy (four had a distal phenotype of MM; one had a phenotype of DMAT and three presented with LGMD2B). It is the first report of genetic confirmed DMAT in China. Mutations c.3112C>T and c.1045dup, may be recurrent mutations in China.
Collapse
Affiliation(s)
- Zhe Zhao
- Department of Neuromuscular Disease, Third Hospital of Hebei Medical University, Shijiazhuang, PR China
| | | | | | | | | | | | | | | |
Collapse
|