1
|
Wang D, Liu XY, He QF, Zheng FZ, Chen L, Zheng Y, Zeng MH, Lin YH, Lin X, Chen HZ, Lin MT, Wang N, Wang ZQ, Lin F. Comprehensive Proteomic Analysis of Dysferlinopathy Unveiling Molecular Mechanisms and Biomarkers Linked to Pathological Progression. CNS Neurosci Ther 2024; 30:e70065. [PMID: 39350328 PMCID: PMC11442333 DOI: 10.1111/cns.70065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/23/2024] [Accepted: 09/13/2024] [Indexed: 10/04/2024] Open
Abstract
AIMS Previous proteomics studies in dysferlinopathy muscle have been limited in scope, often utilizing 2D-electrophoresis and yielding only a small number of differential expression calls. To address this gap, this study aimed to employ high-resolution proteomics to explore the proteomic landscapes of dysferlinopathy and analyze the correlation between muscle pathological changes and alterations in protein expression in muscle biopsies. METHODS We conducted a comprehensive approach to investigate the proteomic profile and disease-associated changes in the muscle tissue proteome from 15 patients with dysferlinopathy, exhibiting varying degrees of dystrophic pathology, alongside age-matched controls. Our methodology encompasses tandem mass tag (TMT)-labeled liquid chromatography-mass spectrometry (LC-MS/MS)-based proteomics, protein-protein interaction (PPI) network analysis, weighted gene co-expression network analysis, and differential expression analysis. Subsequently, we examined the correlation between the expression of key proteins and the clinical characteristics of the patients to identify pathogenic targets associated with DYSF mutations in dysferlinopathy. RESULTS A total of 1600 differentially expressed proteins were identified, with 1321 showing high expression levels and 279 expressed at lower levels. Our investigation yields a molecular profile delineating the altered protein networks in dysferlinopathy-afflicted skeletal muscle, uncovering dysregulation across numerous cellular pathways and molecular processes, including mRNA metabolic processes, regulated exocytosis, immune response, muscle system processes, energy metabolic processes, and calcium transmembrane transport. Moreover, we observe significant associations between the protein expression of ANXA1, ANXA2, ANXA4, ANXA5, LMNA, PYGM, and the extent of histopathologic changes in muscle biopsies from patients with dysferlinopathy, validated through immunoblotting and immunofluorescence assays. CONCLUSIONS Through the aggregation of expression data from dysferlinopathy-impacted muscles exhibiting a range of pathological alterations, we identified multiple key proteins associated with the dystrophic pathology of patients with dysferlinopathy. These findings provide novel insights into the pathogenesis of dysferlinopathy and propose promising targets for future therapeutic endeavors.
Collapse
Affiliation(s)
- Di Wang
- Department of Molecular Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Center for Bioinformatics, National Infrastructures for Translational Medicine, Institute of Clinical Medicine and Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Yi Liu
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Qi-Fang He
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Fu-Ze Zheng
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Long Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Ying Zheng
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Ming-Hui Zeng
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Yu-Hua Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
| | - Xin Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Hai-Zhu Chen
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Min-Ting Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Ning Wang
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Zhi-Qiang Wang
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| | - Feng Lin
- Department of Neurology and Institute of Neurology of First Affiliated Hospital, Institute of Neuroscience, and Fujian Key Laboratory of Molecular Neurology, Fujian Medical University, Fuzhou, China
- Department of Neurology, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China
| |
Collapse
|
2
|
Khodabukus A, Prabhu NK, Roberts T, Buldo M, Detwiler A, Fralish ZD, Kondash ME, Truskey GA, Koves TR, Bursac N. Bioengineered Model of Human LGMD2B Skeletal Muscle Reveals Roles of Intracellular Calcium Overload in Contractile and Metabolic Dysfunction in Dysferlinopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400188. [PMID: 38887849 PMCID: PMC11336985 DOI: 10.1002/advs.202400188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/19/2024] [Indexed: 06/20/2024]
Abstract
Dysferlin is a multi-functional protein that regulates membrane resealing, calcium homeostasis, and lipid metabolism in skeletal muscle. Genetic loss of dysferlin results in limb girdle muscular dystrophy 2B/2R (LGMD2B/2R) and other dysferlinopathies - rare untreatable muscle diseases that lead to permanent loss of ambulation in humans. The mild disease severity in dysferlin-deficient mice and diverse genotype-phenotype relationships in LGMD2B patients have prompted the development of new in vitro models for personalized studies of dysferlinopathy. Here the first 3-D tissue-engineered hiPSC-derived skeletal muscle ("myobundle") model of LGMD2B is described that exhibits compromised contractile function, calcium-handling, and membrane repair, and transcriptomic changes indicative of impaired oxidative metabolism and mitochondrial dysfunction. In response to the fatty acid (FA) challenge, LGMD2B myobundles display mitochondrial deficits and intracellular lipid droplet (LD) accumulation. Treatment with the ryanodine receptor (RyR) inhibitor dantrolene or the dissociative glucocorticoid vamorolone restores LGMD2B contractility, improves membrane repair, and reduces LD accumulation. Lastly, it is demonstrated that chemically induced chronic RyR leak in healthy myobundles phenocopies LGMD2B contractile and metabolic deficit, but not the loss of membrane repair capacity. Together, these results implicate intramyocellular Ca2+ leak as a critical driver of dysferlinopathic phenotype and validate the myobundle system as a platform to study LGMD2B pathogenesis.
Collapse
Affiliation(s)
| | - Neel K. Prabhu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Taylor Roberts
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Meghan Buldo
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Amber Detwiler
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Megan E. Kondash
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Timothy R. Koves
- Duke Molecular Physiology InstituteDuke UniversityDurhamNC27708USA
| | - Nenad Bursac
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
3
|
Jeong HN, Lee TG, Park HJ, Yang Y, Oh SH, Kang SW, Choi YC. Transcriptome analysis of skeletal muscle in dermatomyositis, polymyositis, and dysferlinopathy, using a bioinformatics approach. Front Neurol 2023; 14:1328547. [PMID: 38125829 PMCID: PMC10731051 DOI: 10.3389/fneur.2023.1328547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
Background Polymyositis (PM) and dermatomyositis (DM) are two distinct subgroups of idiopathic inflammatory myopathies. Dysferlinopathy, caused by a dysferlin gene mutation, usually presents in late adolescence with muscle weakness, degenerative muscle changes are often accompanied by inflammatory infiltrates, often resulting in a misdiagnosis as polymyositis. Objective To identify differential biological pathways and hub genes related to polymyositis, dermatomyositis and dysferlinopathy using bioinformatics analysis for understanding the pathomechanisms and providing guidance for therapy development. Methods We analyzed intramuscular ribonucleic acid (RNA) sequencing data from seven dermatomyositis, eight polymyositis, eight dysferlinopathy and five control subjects. Differentially expressed genes (DEGs) were identified by using DESeq2. Enrichment analyses were performed to understand the functions and enriched pathways of DEGs. A protein-protein interaction (PPI) network was constructed, and clarified the gene cluster using the molecular complex detection tool (MCODE) analysis to identify hub genes. Results A total of 1,048, 179 and 3,807 DEGs were detected in DM, PM and dysferlinopathy, respectively. Enrichment analyses revealed that upregulated DEGs were involved in type 1 interferon (IFN1) signaling pathway in DM, antigen processing and presentation of peptide antigen in PM, and cellular response to stimuli in dysferlinopathy. The PPI network and MCODE cluster identified 23 genes related to type 1 interferon signaling pathway in DM, 4 genes (PDIA3, HLA-C, B2M, and TAP1) related to MHC class 1 formation and quality control in PM, and 7 genes (HSPA9, RPTOR, MTOR, LAMTOR1, LAMTOR5, ATP6V0D1, and ATP6V0B) related to cellular response to stress in dysferliniopathy. Conclusion Overexpression of genes related to the IFN1 signaling pathway and major histocompatibility complex (MHC) class I formation was identified in DM and PM, respectively. In dysferlinopathy, overexpression of HSPA9 and the mTORC1 signaling pathway genes was detected.
Collapse
Affiliation(s)
- Ha-Neul Jeong
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Taek Gyu Lee
- Brain Korea 21 PLUS Project for Medical Science, Yonsei University, Seoul, Republic of Korea
| | - Hyung Jun Park
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young Yang
- Research Institute of Women's Disease, Sookmyumg Women's University, Seoul, Republic of Korea
| | - Seung-Hun Oh
- Department of Neurology, CHA Bundang Medical Center, School of Medicine, CHA University, Seongnam-si, Republic of Korea
| | - Seong-Woong Kang
- Department of Rehabilitation Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Young-Chul Choi
- Department of Neurology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Rehabilitation Institute of Neuromuscular Disease, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
4
|
Golding AE, Li W, Blank PS, Cologna SM, Zimmerberg J. Relative quantification of progressive changes in healthy and dysferlin-deficient mouse skeletal muscle proteomes. Muscle Nerve 2023; 68:805-816. [PMID: 37706611 DOI: 10.1002/mus.27975] [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: 11/09/2022] [Revised: 08/22/2023] [Accepted: 08/23/2023] [Indexed: 09/15/2023]
Abstract
INTRODUCTION/AIMS Individuals with dysferlinopathies, a group of genetic muscle diseases, experience delay in the onset of muscle weakness. The cause of this delay and subsequent muscle wasting are unknown, and there are currently no clinical interventions to limit or prevent muscle weakness. To better understand molecular drivers of dysferlinopathies, age-dependent changes in the proteomic profile of skeletal muscle (SM) in wild-type (WT) and dysferlin-deficient mice were identified. METHODS Quadriceps were isolated from 6-, 18-, 42-, and 77-wk-old C57BL/6 (WT, Dysf+/+ ) and BLAJ (Dysf-/- ) mice (n = 3, 2 male/1 female or 1 male/2 female, 24 total). Whole-muscle proteomes were characterized using liquid chromatography-mass spectrometry with relative quantification using TMT10plex isobaric labeling. Principle component analysis was utilized to detect age-dependent proteomic differences over the lifespan of, and between, WT and dysferlin-deficient SM. The biological relevance of proteins with significant variation was established using Ingenuity Pathway Analysis. RESULTS Over 3200 proteins were identified between 6-, 18-, 42-, and 77-wk-old mice. In total, 46 proteins varied in aging WT SM (p < .01), while 365 varied in dysferlin-deficient SM. However, 569 proteins varied between aged-matched WT and dysferlin-deficient SM. Proteins with significant variation in expression across all comparisons followed distinct temporal trends. DISCUSSION Proteins involved in sarcolemma repair and regeneration underwent significant changes in SM over the lifespan of WT mice, while those associated with immune infiltration and inflammation were overly represented over the lifespan of dysferlin-deficient mice. The proteins identified herein are likely to contribute to our overall understanding of SM aging and dysferlinopathy disease progression.
Collapse
Affiliation(s)
- Adriana E Golding
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
- Section on Intracellular Protein Trafficking, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Wenping Li
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois, USA
| | - Paul S Blank
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Stephanie M Cologna
- Department of Chemistry, University of Illinois Chicago, Chicago, Illinois, USA
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
5
|
Ziad F, Cypers G, Phillips M, Vanhoenacker P, Hostens A, Yadavraj S, Lamont D, Robertson T. Adult-onset Alexander disease with unusual inflammatory features and a novel GFAP mutation in two patients. Neuropathol Appl Neurobiol 2023; 49:e12927. [PMID: 37495513 DOI: 10.1111/nan.12927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/19/2023] [Accepted: 07/22/2023] [Indexed: 07/28/2023]
Affiliation(s)
- Fouzia Ziad
- Department of Pathology, Waikato Hospital, Hamilton, New Zealand
| | - Gert Cypers
- Department of Neurology, Onze-Lieve-Vrouwziekenhuis, Aalst, Belgium
| | - Matthew Phillips
- Department of Neurology, Waikato Hospital, Hamilton, New Zealand
| | | | - Arne Hostens
- Department of Neurology, Onze-Lieve-Vrouwziekenhuis, Aalst, Belgium
| | - Satish Yadavraj
- Department of Neurology, Waikato Hospital, Hamilton, New Zealand
| | - Duncan Lamont
- Department of Pathology, Waikato Hospital, Hamilton, New Zealand
| | - Thomas Robertson
- Pathology Queensland, Royal Brisbane and Women's Hospital, Herston, Australia
| |
Collapse
|
6
|
Drescher DG, Drescher MJ, Selvakumar D, Annam NP. Analysis of Dysferlin Direct Interactions with Putative Repair Proteins Links Apoptotic Signaling to Ca 2+ Elevation via PDCD6 and FKBP8. Int J Mol Sci 2023; 24:4707. [PMID: 36902136 PMCID: PMC10002499 DOI: 10.3390/ijms24054707] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/19/2023] [Accepted: 02/24/2023] [Indexed: 03/05/2023] Open
Abstract
Quantitative surface plasmon resonance (SPR) was utilized to determine binding strength and calcium dependence of direct interactions between dysferlin and proteins likely to mediate skeletal muscle repair, interrupted in limb girdle muscular dystrophy type 2B/R2. Dysferlin canonical C2A (cC2A) and C2F/G domains directly interacted with annexin A1, calpain-3, caveolin-3, affixin, AHNAK1, syntaxin-4, and mitsugumin-53, with cC2A the primary target and C2F lesser involved, overall demonstrating positive calcium dependence. Dysferlin C2 pairings alone showed negative calcium dependence in almost all cases. Like otoferlin, dysferlin directly interacted via its carboxy terminus with FKBP8, an anti-apoptotic outer mitochondrial membrane protein, and via its C2DE domain with apoptosis-linked gene (ALG-2/PDCD6), linking anti-apoptosis with apoptosis. Confocal Z-stack immunofluorescence confirmed co-compartmentalization of PDCD6 and FKBP8 at the sarcolemmal membrane. Our evidence supports the hypothesis that prior to injury, dysferlin C2 domains self-interact and give rise to a folded, compact structure as indicated for otoferlin. With elevation of intracellular Ca2+ in injury, dysferlin would unfold and expose the cC2A domain for interaction with annexin A1, calpain-3, mitsugumin 53, affixin, and caveolin-3, and dysferlin would realign from its interactions with PDCD6 at basal calcium levels to interact strongly with FKBP8, an intramolecular rearrangement facilitating membrane repair.
Collapse
Affiliation(s)
- Dennis G. Drescher
- Laboratory of Bio-otology, Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Marian J. Drescher
- Laboratory of Bio-otology, Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Dakshnamurthy Selvakumar
- Laboratory of Bio-otology, Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Neeraja P. Annam
- Laboratory of Bio-otology, Department of Otolaryngology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
7
|
Inhibition of the immunoproteasome modulates innate immunity to ameliorate muscle pathology of dysferlin-deficient BlAJ mice. Cell Death Dis 2022; 13:975. [PMID: 36402750 PMCID: PMC9675822 DOI: 10.1038/s41419-022-05416-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/21/2022]
Abstract
Muscle repair in dysferlinopathies is defective. Although macrophage (Mø)-rich infiltrates are prominent in damaged skeletal muscles of patients with dysferlinopathy, the contribution of the immune system to the disease pathology remains to be fully explored. Numbers of both pro-inflammatory M1 Mø and effector T cells are increased in muscle of dysferlin-deficient BlAJ mice. In addition, symptomatic BlAJ mice have increased muscle production of immunoproteasome. In vitro analyses using bone marrow-derived Mø of BlAJ mice show that immunoproteasome inhibition results in C3aR1 and C5aR1 downregulation and upregulation of M2-associated signaling. Administration of immunoproteasome inhibitor ONX-0914 to BlAJ mice rescues muscle function by reducing muscle infiltrates and fibro-adipogenesis. These findings reveal an important role of immunoproteasome in the progression of muscular dystrophy in BlAJ mouse and suggest that inhibition of immunoproteasome may produce therapeutic benefit in dysferlinopathy.
Collapse
|
8
|
Fernández-Simón E, Lleixà C, Suarez-Calvet X, Diaz-Manera J, Illa I, Gallardo E, de Luna N. Proteasome inhibitors reduce thrombospondin-1 release in human dysferlin-deficient myotubes. BMC Musculoskelet Disord 2020; 21:784. [PMID: 33246442 PMCID: PMC7697384 DOI: 10.1186/s12891-020-03756-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 10/30/2020] [Indexed: 11/16/2022] Open
Abstract
Background Dysferlinopathies are a group of muscle disorders causing muscle weakness and absence or low levels of dysferlin, a type-II transmembrane protein and the causative gene of these dystrophies. Dysferlin is implicated in vesicle fusion, trafficking, and membrane repair. Muscle biopsy of patients with dysferlinopathy is characterized by the presence of inflammatory infiltrates. Studies in the muscle of both human and mouse models of dysferlinopathy suggest dysferlin deficient muscle plays a role in this inflammation by releasing thrombospondin-1. It has also been reported that vitamin D3 treatment enhances dysferlin expression. The ubiquitin-proteasome system recognizes and removes proteins that fail to fold or assemble properly and previous studies suggest that its inhibition could have a therapeutic effect in muscle dystrophies. Here we assessed whether inhibition of the ubiquitin proteasome system prevented degradation of dysferlin in immortalized myoblasts from a patients with two missense mutations in exon 44. Methods To assess proteasome inhibition we treated dysferlin deficient myotubes with EB1089, a vitamin D3 analog, oprozomib and ixazomib. Western blot was performed to analyze the effect of these treatments on the recovery of dysferlin and myogenin expression. TSP-1 was quantified using the enzyme-linked immunosorbent assay to analyze the effect of these drugs on its release. A membrane repair assay was designed to assess the ability of treated myotubes to recover after membrane injury and fusion index was also measured with the different treatments. Data were analyzed using a one-way ANOVA test followed by Tukey post hoc test and analysis of variance. A p ≤ 0.05 was considered statistically significant. Results Treatment with proteasome inhibitors and EB1089 resulted in a trend towards an increase in dysferlin and myogenin expression. Furthermore, EB1089 and proteasome inhibitors reduced the release of TSP-1 in myotubes. However, no effect was observed on the repair of muscle membrane after injury. Conclusions Our findings indicate that the ubiquitin-proteasome system might not be the main mechanism of mutant dysferlin degradation. However, its inhibition could help to improve muscle inflammation by reducing TSP-1 release. Supplementary Information The online version contains supplementary material available at 10.1186/s12891-020-03756-7.
Collapse
Affiliation(s)
- Esther Fernández-Simón
- Neuromuscular Diseases group. Institut de Recerca Hospital de Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autónoma de Barcelona, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
| | - Cinta Lleixà
- Neuromuscular Diseases group. Institut de Recerca Hospital de Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autónoma de Barcelona, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain
| | - Xavier Suarez-Calvet
- Neuromuscular Diseases group. Institut de Recerca Hospital de Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autónoma de Barcelona, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Jordi Diaz-Manera
- Neuromuscular Diseases group. Institut de Recerca Hospital de Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autónoma de Barcelona, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain.,Department of Neurology, Neuromuscular Diseases Unit, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Isabel Illa
- Neuromuscular Diseases group. Institut de Recerca Hospital de Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autónoma de Barcelona, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain.,Department of Neurology, Neuromuscular Diseases Unit, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eduard Gallardo
- Neuromuscular Diseases group. Institut de Recerca Hospital de Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autónoma de Barcelona, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain.
| | - Noemí de Luna
- Neuromuscular Diseases group. Institut de Recerca Hospital de Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Universitat Autónoma de Barcelona, c/Sant Antoni Mª Claret 167, 08025, Barcelona, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Valencia, Spain.
| |
Collapse
|
9
|
Evans S, Weinheimer CJ, Kovacs A, Williams JW, Randolph GJ, Jiang W, Barger PM, Mann DL. Ischemia reperfusion injury provokes adverse left ventricular remodeling in dysferlin-deficient hearts through a pathway that involves TIRAP dependent signaling. Sci Rep 2020; 10:14129. [PMID: 32839504 PMCID: PMC7445276 DOI: 10.1038/s41598-020-71079-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/04/2020] [Indexed: 12/17/2022] Open
Abstract
Cardiac myocytes have multiple cell autonomous mechanisms that facilitate stabilization and repair of damaged sarcolemmal membranes following myocardial injury. Dysferlin is a protein which facilitates membrane repair by promoting membrane resealing. Although prior studies have shown that dysferlin-deficient (Dysf-/-) mouse hearts have an impaired recovery from acute ischemia/reperfusion (I/R) injury ex vivo, the role of dysferlin in mediating the recovery from myocardial injury in vivo is unknown. Here we show that Dysf-/- mice develop adverse LV remodeling following I/R injury secondary to the collateral damage from sustained myocardial inflammation within the infarct zone. Backcrossing Dysf-/- mice with mice lacking signaling through the Toll-Interleukin 1 Receptor Domain-Containing Adaptor Protein (Tirap-/-), attenuated inflammation and abrogated adverse LV remodeling following I/R injury. Subsequent studies using Poloxamer 188 (P188), a membrane resealing reagent, demonstrated that P188 did not attenuate inflammation nor prevent adverse LV remodeling in Dysf-/- mice following I/R injury. Viewed together these studies reveal a previously unappreciated role for the importance of membrane sealing and the resolution of inflammation following myocardial injury.
Collapse
Affiliation(s)
- Sarah Evans
- Center for Cardiovascular Research, Cardiovascular Division, Division of Cardiology, Washington University School of Medicine, 660 S. Euclid Ave,, Campus Box 8086, St. Louis, MO, 63110, USA
| | - Carla J Weinheimer
- Center for Cardiovascular Research, Cardiovascular Division, Division of Cardiology, Washington University School of Medicine, 660 S. Euclid Ave,, Campus Box 8086, St. Louis, MO, 63110, USA
| | - Attila Kovacs
- Center for Cardiovascular Research, Cardiovascular Division, Division of Cardiology, Washington University School of Medicine, 660 S. Euclid Ave,, Campus Box 8086, St. Louis, MO, 63110, USA
| | - Jesse W Williams
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gwendalyn J Randolph
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Wenlong Jiang
- Center for Cardiovascular Research, Cardiovascular Division, Division of Cardiology, Washington University School of Medicine, 660 S. Euclid Ave,, Campus Box 8086, St. Louis, MO, 63110, USA
| | - Philip M Barger
- Center for Cardiovascular Research, Cardiovascular Division, Division of Cardiology, Washington University School of Medicine, 660 S. Euclid Ave,, Campus Box 8086, St. Louis, MO, 63110, USA
| | - Douglas L Mann
- Center for Cardiovascular Research, Cardiovascular Division, Division of Cardiology, Washington University School of Medicine, 660 S. Euclid Ave,, Campus Box 8086, St. Louis, MO, 63110, USA.
| |
Collapse
|
10
|
Wang P, Xu L, Gao Y, Han R. BEON: A Functional Fluorescence Reporter for Quantification and Enrichment of Adenine Base-Editing Activity. Mol Ther 2020; 28:1696-1705. [PMID: 32353322 PMCID: PMC7335737 DOI: 10.1016/j.ymthe.2020.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 02/12/2020] [Accepted: 04/09/2020] [Indexed: 12/11/2022] Open
Abstract
Adenine base editor (ABE) is a new generation of genome-editing technology through fusion of Cas9 nickase with an evolved E. coli TadA (TadA∗) and holds great promise as novel genome-editing therapeutics for treating genetic disorders. ABEs can directly convert A-T to G-C in specific genomic DNA targets without introducing double-strand breaks (DSBs). We recently showed that computer program-assisted analysis of Sanger sequencing traces can be used as a low-cost and rapid alternative of deep sequencing to assess base-editing outcomes. Here we developed a rapid fluorescence-based reporter assay (Base Editing ON [BEON]) to quantify ABE efficiency. The assay relies on the restoration of the downstream green fluorescent protein (GFP) in ABE-mediated editing of a stop codon located within the guide RNA (gRNA). We showed that this assay can be used to screen for effective ABE variants, characterize the protospacer adjacent motif (PAM) requirement of a novel NNG-targeting ABE based on ScCas9, and enrich for edited cells. Finally, we demonstrated that the reporter assay allowed us to assess the feasibility of ABE editing to correct point mutations associated with dysferlinopathy. Taken together, the BEON assay would facilitate and simplify the studies with ABEs.
Collapse
Affiliation(s)
- Peipei Wang
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Li Xu
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Yandi Gao
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Renzhi Han
- Department of Surgery, Davis Heart and Lung Research Institute, Biomedical Sciences Graduate Program, Biophysics Graduate Program, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
11
|
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
|
12
|
Urao N, Mirza RE, Corbiere TF, Hollander Z, Borchers CH, Koh TJ. Thrombospondin-1 and disease progression in dysferlinopathy. Hum Mol Genet 2018; 26:4951-4960. [PMID: 29206970 DOI: 10.1093/hmg/ddx378] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/05/2017] [Indexed: 01/30/2023] Open
Abstract
The purpose of this study was to determine whether thrombospondin (TSP)-1 promotes macrophage activity and disease progression in dysferlinopathy. First, we found that levels of TSP-1 are elevated in blood of non-ambulant dysferlinopathy patients compared with ambulant patients and healthy controls, supporting the idea that TSP-1 levels are correlated with disease progression. We then crossed dysferlinopathic BlaJ mice with TSP-1 knockout mice and assessed disease progression longitudinally with magnetic resonance imaging (MRI). In these mice, deletion of TSP-1 ameliorated loss in volume and mass of the moderately affected gluteal muscle but not of the severely affected psoas muscle. T2 MRI parameters revealed that loss of TSP-1 modestly inhibited inflammation only in gluteal muscle of male mice. Histological assessment indicated that deletion of TSP-1 reduced inflammatory cell infiltration of muscle fibers, but only early in disease progression. In addition, flow cytometry analysis revealed that, in males, TSP-1 knockout reduced macrophage infiltration and phagocytic activity, which is consistent with TSP-1-enhanced phagocytosis and pro-inflammatory cytokine induction in cultured macrophages. In summary, TSP-1 appears to play an accessory role in modulating Mp activity in BlaJ mice in a gender, age and muscle-dependent manner, but is unlikely a primary driver of disease progression of dysferlinopathy.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA.,Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rita E Mirza
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Thomas F Corbiere
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Zsuzsanna Hollander
- PROOF Center of Excellence, Vancouver, BC, Canada.,UBC James Hogg Research Centre, Vancouver, BC, Canada
| | - Christoph H Borchers
- University of Victoria - Genome British Columbia Proteomics Centre, University of Victoria, Victoria, BC, Canada.,Department of Biochemistry and Microbiology, University of Victoria, Victoria, BC, Canada.,Proteomics Centre, Segal Cancer Centre, Lady Davis Institute, Jewish General Hospital, McGill University, Montreal, QC, Canada.,Gerald Bronfman Department of Oncology, Jewish General Hospital, Montreal, QC, Canada
| | - Timothy J Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA.,Center for Wound Healing and Tissue Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
13
|
Potter RA, Griffin DA, Sondergaard PC, Johnson RW, Pozsgai ER, Heller KN, Peterson EL, Lehtimäki KK, Windish HP, Mittal PJ, Albrecht DE, Mendell JR, Rodino-Klapac LR. Systemic Delivery of Dysferlin Overlap Vectors Provides Long-Term Gene Expression and Functional Improvement for Dysferlinopathy. Hum Gene Ther 2018; 29:749-762. [PMID: 28707952 PMCID: PMC6066196 DOI: 10.1089/hum.2017.062] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 07/12/2017] [Indexed: 01/07/2023] Open
Abstract
Dysferlinopathies comprise a family of disorders caused by mutations in the dysferlin (DYSF) gene, leading to a progressive dystrophy characterized by chronic muscle fiber loss, fat replacement, and fibrosis. To correct the underlying histopathology and function, expression of full-length DYSF is required. Dual adeno-associated virus vectors have been developed, defined by a region of homology, to serve as a substrate for reconstitution of the full 6.5 kb dysferlin cDNA. Previous work studied the efficacy of this treatment through intramuscular and regional delivery routes. To maximize clinical efficacy, dysferlin-deficient mice were treated systemically to target all muscles through the vasculature for efficacy and safety studies. Mice were evaluated at multiple time points between 4 and 13 months post treatment for dysferlin expression and functional improvement using magnetic resonance imaging and magnetic resonance spectroscopy and membrane repair. A systemic dose of 6 × 1012 vector genomes resulted in widespread gene expression in the muscles. Treated muscles showed a significant decrease in central nucleation, collagen deposition, and improvement of membrane repair to wild-type levels. Treated gluteus muscles were significantly improved compared to placebo-treated muscles and were equivalent to wild type in volume, intra- and extramyocellular lipid accumulation, and fat percentage using magnetic resonance imaging and magnetic resonance spectroscopy. Dual-vector treatment allows for production of full-length functional dysferlin with no toxicity. This confirms previous safety data and validates translation of systemic gene delivery for dysferlinopathy patients.
Collapse
Affiliation(s)
- Rachael A. Potter
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Danielle A. Griffin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Patricia C. Sondergaard
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Ryan W. Johnson
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Eric R. Pozsgai
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio; The Ohio State University, Columbus, Ohio
| | - Kristin N. Heller
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | - Ellyn L. Peterson
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
| | | | | | | | | | - Jerry R. Mendell
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics and Neurology, The Ohio State University, Columbus, Ohio; The Ohio State University, Columbus, Ohio
| | - Louise R. Rodino-Klapac
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio
- Department of Pediatrics and Neurology, The Ohio State University, Columbus, Ohio; The Ohio State University, Columbus, Ohio
- Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, Ohio; The Ohio State University, Columbus, Ohio
| |
Collapse
|
14
|
Collier AF, Gumerson J, Lehtimäki K, Puoliväli J, Jones JW, Kane MA, Manne S, O'Neill A, Windish HP, Ahtoniemi T, Williams BA, Albrecht DE, Bloch RJ. Effect of Ibuprofen on Skeletal Muscle of Dysferlin-Null Mice. J Pharmacol Exp Ther 2017; 364:409-419. [PMID: 29284661 DOI: 10.1124/jpet.117.244244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/21/2017] [Indexed: 12/19/2022] Open
Abstract
Ibuprofen, a nonsteroidal anti-inflammatory drug, and nitric oxide (NO) donors have been reported to reduce the severity of muscular dystrophies in mice associated with the absence of dystrophin or α-sarcoglycan, but their effects on mice that are dystrophic due to the absence of dysferlin have not been examined. We have tested ibuprofen, as well as isosorbide dinitrate (ISDN), a NO donor, to learn whether used alone or together they protect dysferlin-null muscle in A/J mice from large strain injury (LSI) induced by a series of high strain lengthening contractions. Mice were maintained on chow containing ibuprofen and ISDN for 4 weeks. They were then subjected to LSI and maintained on the drugs for 3 additional days. We measured loss of torque immediately following injury and at day 3 postinjury, fiber necrosis, and macrophage infiltration at day 3 postinjury, and serum levels of the drugs at the time of euthanasia. Loss of torque immediately after injury was not altered by the drugs. However, the torque on day 3 postinjury significantly decreased as a function of ibuprofen concentration in the serum (range, 0.67-8.2 µg/ml), independent of ISDN. The effects of ISDN on torque loss at day 3 postinjury were not significant. In long-term studies of dysferlinopathic BlAJ mice, lower doses of ibuprofen had no effects on muscle morphology, but reduced treadmill running by 40%. Our results indicate that ibuprofen can have deleterious effects on dysferlin-null muscle and suggest that its use at pharmacological doses should be avoided by individuals with dysferlinopathies.
Collapse
Affiliation(s)
- Alyssa F Collier
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Jessica Gumerson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Kimmo Lehtimäki
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Jukka Puoliväli
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Jace W Jones
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Maureen A Kane
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Sankeerth Manne
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Andrea O'Neill
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Hillarie P Windish
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Toni Ahtoniemi
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Bradley A Williams
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Douglas E Albrecht
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Robert J Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| |
Collapse
|
15
|
Defour A, Medikayala S, Van der Meulen JH, Hogarth MW, Holdreith N, Malatras A, Duddy W, Boehler J, Nagaraju K, Jaiswal JK. Annexin A2 links poor myofiber repair with inflammation and adipogenic replacement of the injured muscle. Hum Mol Genet 2017; 26:1979-1991. [PMID: 28334824 DOI: 10.1093/hmg/ddx065] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 02/17/2017] [Indexed: 01/12/2023] Open
Abstract
Repair of skeletal muscle after sarcolemmal damage involves dysferlin and dysferlin-interacting proteins such as annexins. Mice and patient lacking dysferlin exhibit chronic muscle inflammation and adipogenic replacement of the myofibers. Here, we show that similar to dysferlin, lack of annexin A2 (AnxA2) also results in poor myofiber repair and progressive muscle weakening with age. By longitudinal analysis of AnxA2-deficient muscle we find that poor myofiber repair due to the lack of AnxA2 does not result in chronic inflammation or adipogenic replacement of the myofibers. Further, deletion of AnxA2 in dysferlin deficient mice reduced muscle inflammation, adipogenic replacement of myofibers, and improved muscle function. These results identify multiple roles of AnxA2 in muscle repair, which includes facilitating myofiber repair, chronic muscle inflammation and adipogenic replacement of dysferlinopathic muscle. It also identifies inhibition of AnxA2-mediated inflammation as a novel therapeutic avenue for treating muscle loss in dysferlinopathy.
Collapse
Affiliation(s)
- Aurelia Defour
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Sushma Medikayala
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Jack H Van der Meulen
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Marshall W Hogarth
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Nicholas Holdreith
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Apostolos Malatras
- Center for Research in Myology 75013, Sorbonne Universités, UPMC University Paris 06, INSERM UMRS975, CNRS FRE3617, GH Pitié Salpêtrière, Paris 13, Paris, France
| | - William Duddy
- Center for Research in Myology 75013, Sorbonne Universités, UPMC University Paris 06, INSERM UMRS975, CNRS FRE3617, GH Pitié Salpêtrière, Paris 13, Paris, France
- Northern Ireland Centre for Stratified Medicine, Altnagelvin Hospital Campus, Ulster University, Londonderry, Northern Ireland, BT52 1SJ UK
| | - Jessica Boehler
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20052 USA
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Health System, Washington, DC 20010, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC, 20052 USA
| |
Collapse
|
16
|
Lukyanenko V, Muriel JM, Bloch RJ. Coupling of excitation to Ca 2+ release is modulated by dysferlin. J Physiol 2017; 595:5191-5207. [PMID: 28568606 DOI: 10.1113/jp274515] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Accepted: 05/16/2017] [Indexed: 12/16/2022] Open
Abstract
KEY POINTS Dysferlin, the protein missing in limb girdle muscular dystrophy 2B and Miyoshi myopathy, concentrates in transverse tubules of skeletal muscle, where it stabilizes voltage-induced Ca2+ transients against loss after osmotic shock injury (OSI). Local expression of dysferlin in dysferlin-null myofibres increases transient amplitude to control levels and protects them from loss after OSI. Inhibitors of ryanodine receptors (RyR1) and L-type Ca2+ channels protect voltage-induced Ca2+ transients from loss; thus both proteins play a role in injury in dysferlin's absence. Effects of Ca2+ -free medium and S107, which inhibits SR Ca2+ leak, suggest the SR as the primary source of Ca2+ responsible for the loss of the Ca2+ transient upon injury. Ca2+ waves were induced by OSI and suppressed by exogenous dysferlin. We conclude that dysferlin prevents injury-induced SR Ca2+ leak. ABSTRACT Dysferlin concentrates in the transverse tubules of skeletal muscle and stabilizes Ca2+ transients when muscle fibres are subjected to osmotic shock injury (OSI). We show here that voltage-induced Ca2+ transients elicited in dysferlin-null A/J myofibres were smaller than control A/WySnJ fibres. Regional expression of Venus-dysferlin chimeras in A/J fibres restored the full amplitude of the Ca2+ transients and protected against OSI. We also show that drugs that target ryanodine receptors (RyR1: dantrolene, tetracaine, S107) and L-type Ca2+ channels (LTCCs: nifedipine, verapamil, diltiazem) prevented the decrease in Ca2+ transients in A/J fibres following OSI. Diltiazem specifically increased transients by ∼20% in uninjured A/J fibres, restoring them to control values. The fact that both RyR1s and LTCCs were involved in OSI-induced damage suggests that damage is mediated by increased Ca2+ leak from the sarcoplasmic reticulum (SR) through the RyR1. Congruent with this, injured A/J fibres produced Ca2+ sparks and Ca2+ waves. S107 (a stabilizer of RyR1-FK506 binding protein coupling that reduces Ca2+ leak) or local expression of Venus-dysferlin prevented OSI-induced Ca2+ waves. Our data suggest that dysferlin modulates SR Ca2+ release in skeletal muscle, and that in its absence OSI causes increased RyR1-mediated Ca2+ leak from the SR into the cytoplasm.
Collapse
Affiliation(s)
- Valeriy Lukyanenko
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Joaquin M Muriel
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Robert J Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, MD, USA
| |
Collapse
|
17
|
Limb-Girdle Muscular Dystrophy 2B and Miyoshi Presentations of Dysferlinopathy. Am J Med Sci 2017; 353:484-491. [DOI: 10.1016/j.amjms.2016.05.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Revised: 05/16/2016] [Accepted: 05/17/2016] [Indexed: 11/20/2022]
|
18
|
Blazek AD, Paleo BJ, Weisleder N. Plasma Membrane Repair: A Central Process for Maintaining Cellular Homeostasis. Physiology (Bethesda) 2016; 30:438-48. [PMID: 26525343 DOI: 10.1152/physiol.00019.2015] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Plasma membrane repair is a conserved cellular response mediating active resealing of membrane disruptions to maintain homeostasis and prevent cell death and progression of multiple diseases. Cell membrane repair repurposes mechanisms from various cellular functions, including vesicle trafficking, exocytosis, and endocytosis, to mend the broken membrane. Recent studies increased our understanding of membrane repair by establishing the molecular machinery contributing to membrane resealing. Here, we review some of the key proteins linked to cell membrane repair.
Collapse
Affiliation(s)
- Alisa D Blazek
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Brian J Paleo
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| | - Noah Weisleder
- Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, Ohio
| |
Collapse
|
19
|
Cárdenas AM, González-Jamett AM, Cea LA, Bevilacqua JA, Caviedes P. Dysferlin function in skeletal muscle: Possible pathological mechanisms and therapeutical targets in dysferlinopathies. Exp Neurol 2016; 283:246-54. [PMID: 27349407 DOI: 10.1016/j.expneurol.2016.06.026] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 12/18/2022]
Abstract
Mutations in the dysferlin gene are linked to a group of muscular dystrophies known as dysferlinopathies. These myopathies are characterized by progressive atrophy. Studies in muscle tissue from dysferlinopathy patients or dysferlin-deficient mice point out its importance in membrane repair. However, expression of dysferlin homologous proteins that restore sarcolemma repair function in dysferlinopathy animal models fail to arrest muscle wasting, therefore suggesting that dysferlin plays other critical roles in muscle function. In the present review, we discuss dysferlin functions in the skeletal muscle, as well as pathological mechanisms related to dysferlin mutations. Particular focus is presented related the effect of dysferlin on cell membrane related function, which affect its repair, vesicle trafficking, as well as Ca(2+) homeostasis. Such mechanisms could provide accessible targets for pharmacological therapies.
Collapse
Affiliation(s)
- Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.
| | - Arlek M González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Luis A Cea
- Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Jorge A Bevilacqua
- Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Pablo Caviedes
- Programa de Farmacología Molecular y Clinica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
20
|
Demonbreun AR, Allen MV, Warner JL, Barefield DY, Krishnan S, Swanson KE, Earley JU, McNally EM. Enhanced Muscular Dystrophy from Loss of Dysferlin Is Accompanied by Impaired Annexin A6 Translocation after Sarcolemmal Disruption. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:1610-22. [PMID: 27070822 DOI: 10.1016/j.ajpath.2016.02.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 01/20/2016] [Accepted: 02/11/2016] [Indexed: 02/03/2023]
Abstract
Dysferlin is a membrane-associated protein implicated in membrane resealing; loss of dysferlin leads to muscular dystrophy. We examined the same loss-of-function Dysf mutation in two different mouse strains, 129T2/SvEmsJ (Dysf(129)) and C57BL/6J (Dysf(B6)). Although there are many genetic differences between these two strains, we focused on polymorphisms in Anxa6 because these variants were previously associated with modifying a pathologically distinct form of muscular dystrophy and increased the production of a truncated annexin A6 protein. Dysferlin deficiency in the C57BL/6J background was associated with increased Evan's Blue dye uptake into muscle and increased serum creatine kinase compared to the 129T2/SvEmsJ background. In the C57BL/6J background, dysferlin loss was associated with enhanced pathologic severity, characterized by decreased mean fiber cross-sectional area, increased internalized nuclei, and increased fibrosis, compared to that in Dysf(129) mice. Macrophage infiltrate was also increased in Dysf(B6) muscle. High-resolution imaging of live myofibers demonstrated that fibers from Dysf(B6) mice displayed reduced translocation of full-length annexin A6 to the site of laser-induced sarcolemmal disruption compared to Dysf(129) myofibers, and impaired translocation of annexin A6 associated with impaired resealing of the sarcolemma. These results provide one mechanism by which the C57BL/6J background intensifies dysferlinopathy, giving rise to a more severe form of muscular dystrophy in the Dysf(B6) mouse model through increased membrane leak and inflammation.
Collapse
Affiliation(s)
| | - Madison V Allen
- Center for Genetic Medicine, Northwestern University, Chicago, Illinois
| | - James L Warner
- Center for Genetic Medicine, Northwestern University, Chicago, Illinois
| | - David Y Barefield
- Center for Genetic Medicine, Northwestern University, Chicago, Illinois
| | - Swathi Krishnan
- Department of Medicine, The University of Chicago, Chicago, Illinois
| | - Kaitlin E Swanson
- Department of Pathology, The University of Chicago, Chicago, Illinois
| | - Judy U Earley
- Center for Genetic Medicine, Northwestern University, Chicago, Illinois
| | | |
Collapse
|
21
|
Urao N, Mirza RE, Heydemann A, Garcia J, Koh TJ. Thrombospondin-1 levels correlate with macrophage activity and disease progression in dysferlin deficient mice. Neuromuscul Disord 2016; 26:240-51. [PMID: 26927626 DOI: 10.1016/j.nmd.2016.01.002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 11/24/2015] [Accepted: 01/14/2016] [Indexed: 10/22/2022]
Abstract
Dysferlinopathy is associated with accumulation of thrombospondin (TSP)-1 and macrophages, both of which may contribute to the pathogenesis of the disease. The purpose of this study was to determine whether TSP-1 levels can predict macrophage activity and disease progression in dysferlin deficient BlaJ mice, focusing on the early disease process. In 3 month-old BlaJ mice, muscle TSP-1 levels exhibited strong positive correlations with both accumulation of F4/80hi macrophages and with their in vivo phagocytic activity in psoas muscles as measured by magnetic resonance imaging and flow cytometry. Muscle TSP-1 levels also exhibited a strong negative correlation with muscle mass and strong positive correlations with histological measurements of muscle fiber infiltration and regeneration. Over the course of disease progression from 3 to 12 months of age, muscle TSP-1 levels showed more complicated relationships with macrophage activity and an inverse relationship with muscle mass. Importantly, blood TSP-1 levels showed strong correlations with macrophage activity and muscle degeneration, particularly early in disease progression in BlaJ mice. These data indicate that TSP-1 may contribute to a destructive macrophage response in dysferlinopathy and pose the intriguing possibility that TSP-1 levels may serve as a biomarker for disease progression.
Collapse
Affiliation(s)
- Norifumi Urao
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA; Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Rita E Mirza
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Ahlke Heydemann
- Department of Physiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jesus Garcia
- Department of Physiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Timothy J Koh
- Department of Kinesiology and Nutrition, University of Illinois at Chicago, Chicago, IL 60612, USA; Center for Tissue Repair and Regeneration, University of Illinois at Chicago, Chicago, IL 60612, USA.
| |
Collapse
|
22
|
Kanagawa M, Lu Z, Ito C, Matsuda C, Miyake K, Toda T. Contribution of dysferlin deficiency to skeletal muscle pathology in asymptomatic and severe dystroglycanopathy models: generation of a new model for Fukuyama congenital muscular dystrophy. PLoS One 2014; 9:e106721. [PMID: 25198651 PMCID: PMC4157776 DOI: 10.1371/journal.pone.0106721] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 08/01/2014] [Indexed: 11/18/2022] Open
Abstract
Defects in dystroglycan glycosylation are associated with a group of muscular dystrophies, termed dystroglycanopathies, that include Fukuyama congenital muscular dystrophy (FCMD). It is widely believed that abnormal glycosylation of dystroglycan leads to disease-causing membrane fragility. We previously generated knock-in mice carrying a founder retrotransposal insertion in fukutin, the gene responsible for FCMD, but these mice did not develop muscular dystrophy, which hindered exploring therapeutic strategies. We hypothesized that dysferlin functions may contribute to muscle cell viability in the knock-in mice; however, pathological interactions between glycosylation abnormalities and dysferlin defects remain unexplored. To investigate contributions of dysferlin deficiency to the pathology of dystroglycanopathy, we have crossed dysferlin-deficient dysferlin(sjl/sjl) mice to the fukutin-knock-in fukutin(Hp/-) and Large-deficient Largemyd/myd mice, which are phenotypically distinct models of dystroglycanopathy. The fukutin(Hp/-) mice do not show a dystrophic phenotype; however, (dysferlin(sjl/sjl): fukutin(Hp/-)) mice showed a deteriorated phenotype compared with (dysferlinsjl/sjl: fukutin(Hp/+)) mice. These data indicate that the absence of functional dysferlin in the asymptomatic fukutin(Hp/-) mice triggers disease manifestation and aggravates the dystrophic phenotype. A series of pathological analyses using double mutant mice for Large and dysferlin indicate that the protective effects of dysferlin appear diminished when the dystrophic pathology is severe and also may depend on the amount of dysferlin proteins. Together, our results show that dysferlin exerts protective effects on the fukutin(Hp/-) FCMD mouse model, and the (dysferlin(sjl/sjl): fukutin(Hp/-)) mice will be useful as a novel model for a recently proposed antisense oligonucleotide therapy for FCMD.
Collapse
Affiliation(s)
- Motoi Kanagawa
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Zhongpeng Lu
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Chiyomi Ito
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Chie Matsuda
- Biomedical Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Katsuya Miyake
- Department of Histology and Cell Biology, School of Medicine, Kagawa University, Ikenobe, Miki, Kagawa, Japan
| | - Tatsushi Toda
- Division of Neurology/Molecular Brain Science, Kobe University Graduate School of Medicine, Kobe, Japan
- * E-mail:
| |
Collapse
|
23
|
Influence of immune responses in gene/stem cell therapies for muscular dystrophies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:818107. [PMID: 24959590 PMCID: PMC4052166 DOI: 10.1155/2014/818107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/07/2014] [Accepted: 04/30/2014] [Indexed: 02/06/2023]
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of diseases, caused by mutations in different components of sarcolemma, extracellular matrix, or enzymes. Inflammation and innate or adaptive immune response activation are prominent features of MDs. Various therapies under development are directed toward rescuing the dystrophic muscle damage using gene transfer or cell therapy. Here we discussed current knowledge about involvement of immune system responses to experimental therapies in MDs.
Collapse
|
24
|
Grounds MD, Terrill JR, Radley-Crabb HG, Robertson T, Papadimitriou J, Spuler S, Shavlakadze T. Lipid accumulation in dysferlin-deficient muscles. THE AMERICAN JOURNAL OF PATHOLOGY 2014; 184:1668-76. [PMID: 24685690 DOI: 10.1016/j.ajpath.2014.02.005] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Revised: 02/21/2014] [Accepted: 02/25/2014] [Indexed: 02/01/2023]
Abstract
Dysferlin is a membrane associated protein involved in vesicle trafficking and fusion. Defects in dysferlin result in limb-girdle muscular dystrophy type 2B and Miyoshi myopathy in humans and myopathy in A/J(dys-/-) and BLAJ mice, but the pathomechanism of the myopathy is not understood. Oil Red O staining showed many lipid droplets within the psoas and quadriceps muscles of dysferlin-deficient A/J(dys-/-) mice aged 8 and 12 months, and lipid droplets were also conspicuous within human myofibers from patients with dysferlinopathy (but not other myopathies). Electron microscopy of 8-month-old A/J(dys-/-) psoas muscles confirmed lipid droplets within myofibers and showed disturbed architecture of myofibers. In addition, the presence of many adipocytes was confirmed, and a possible role for dysferlin in adipocytes is suggested. Increased expression of mRNA for a gene involved in early lipogenesis, CCAAT/enhancer binding protein-δ, in 3-month-old A/J(dys-/-) quadriceps (before marked histopathology is evident), indicates early induction of lipogenesis/adipogenesis within dysferlin-deficient muscles. Similar results were seen for dysferlin-deficient BLAJ mice. These novel observations of conspicuous intermyofibrillar lipid and progressive adipocyte replacement in dysferlin-deficient muscles present a new focus for investigating the mechanisms that result in the progressive decline of muscle function in dysferlinopathies.
Collapse
Affiliation(s)
- Miranda D Grounds
- Schools of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Australia.
| | - Jessica R Terrill
- Schools of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Australia
| | - Hannah G Radley-Crabb
- Schools of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Australia; CHIRI Biosciences Research Precinct, School of Biomedical Sciences, Curtin University, Perth, Australia
| | - Terry Robertson
- Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - John Papadimitriou
- Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - Simone Spuler
- Muscle Research Unit, Experimental and Clinical Research Center, Berlin, Germany
| | - Tea Shavlakadze
- Schools of Anatomy, Physiology and Human Biology, University of Western Australia, Perth, Australia
| |
Collapse
|
25
|
Smith RC, Lin BK. Myostatin inhibitors as therapies for muscle wasting associated with cancer and other disorders. Curr Opin Support Palliat Care 2013; 7:352-60. [PMID: 24157714 PMCID: PMC3819341 DOI: 10.1097/spc.0000000000000013] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW This review summarizes recent progress in the development of myostatin inhibitors for the treatment of muscle wasting disorders. It also focuses on findings in myostatin biology that may have implications for the development of antimyostatin therapies. RECENT FINDINGS There has been progress in evaluating antimyostatin therapies in animal models of muscle wasting disorders. Some programs have progressed into clinical development with initial results showing positive impact on muscle volume.In normal mice myostatin deficiency results in enlarged muscles with increased total force but decreased specific force (total force/total mass). An increase in myofibrillar protein synthesis without concomitant satellite cell proliferation and fusion leads to muscle hypertrophy with unchanged myonuclear number. A specific force reduction is not observed when atrophied muscle, the predominant therapeutic target of myostatin inhibitor therapy, is made myostatindeficient.Myostatin has been shown to be expressed by a number of tumor cell lines in mice and man. SUMMARY Myostatin inhibition remains a promising therapeutic strategy for a range of muscle wasting disorders.
Collapse
Affiliation(s)
- Rosamund C Smith
- aBiotechnology Discovery Research bOncology Business Unit, Eli Lilly and Company
| | | |
Collapse
|