1
|
Tammineni ER, Manno C, Oza G, Figueroa L. Skeletal muscle disorders as risk factors for type 2 diabetes. Mol Cell Endocrinol 2025; 599:112466. [PMID: 39848431 PMCID: PMC11886953 DOI: 10.1016/j.mce.2025.112466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/27/2024] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
The incidence and prevalence of muscular disorders and of type 2 diabetes (T2D) is increasing and both represent highly significant healthcare problems, both economically and compromising quality of life. Interestingly, skeletal muscle dysfunction and T2D share some commonalities including dysregulated glucose homeostasis, increased oxidative stress, dyslipidemia, and cytokine alterations. Several lines of evidence have hinted to a relationship between skeletal muscle dysfunction and T2D. For instance, T2D affects skeletal muscle morphology, functionality, and overall health through altered protein metabolism, impaired mitochondrial function, and ultimately cell viability. Conversely, humans suffering from myopathies and their experimental models demonstrated increased incidence of T2D through altered muscle glucose disposal function due to abnormal calcium homeostasis, compromised mitochondrial function, dyslipidemia, increased inflammatory cytokines and fiber size alterations and disproportions. Lifestyle modifications are essential for improving and maintaining mobility and metabolic health in individuals suffering from myopathies along with T2D. In this review, we updated current literature evidence on clinical incidence of T2D in inflammatory, mitochondrial, metabolic myopathies, and muscular dystrophies and further discussed the molecular basis of these skeletal muscle disorders leading to T2D.
Collapse
Affiliation(s)
| | - Carlo Manno
- Department of Physiology and Biophysics, Rush University, Chicago, USA
| | - Goldie Oza
- Centro de Investigación y Desarrollo Tecnológico en Electroquímica S. C., Queretaro, Mexico
| | - Lourdes Figueroa
- Department of Physiology and Biophysics, Rush University, Chicago, USA
| |
Collapse
|
2
|
Lauletta A, Allenbach Y, Béhin A, Evangelista T, Léonard-Louis S, Garibaldi M, Benveniste O. High prevalence of facioscapulohumeral muscular dystrophy (FSHD) and inflammatory myopathies association: Is there an interplay? J Neurol Sci 2025; 470:123400. [PMID: 39855012 DOI: 10.1016/j.jns.2025.123400] [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: 06/09/2024] [Revised: 12/19/2024] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
INTRODUCTION Certain types of muscular dystrophy (MD), notably facioscapulohumeral muscular dystrophy (FSHD), exhibit muscle fiber necrosis with regeneration and a nonspecific inflammatory process. Although rare, the coexistence of MDs and autoimmune myositis has been observed. We hypothesized that, in some circumstances, FSHD may predispose individuals to myositis through muscle damage-induced autoantigen overexpression, contributing to an autoimmune response. METHODS We conducted a retrospective analysis of patient data from neuromuscular disease centers in France and Italy between September 2012 and May 2024. Clinical, immunological, and myopathological features of 1750 myositis patients were comprehensively reviewed. RESULTS Five patients were identified with both FSHD and IIM. Two patients were first diagnosed with FSHD and later developed IIM, while two others initially had IIM followed by an FSHD diagnosis. The fifth patient received simultaneous diagnoses of both conditions. The prevalence of FSHD in the IIM cohort was 1/350, and the prevalence of IIM in the FSHD cohort was 1/40 (p < 0.0001). DISCUSSION Our study showed a high prevalence of FSHD and IIM association compared to the general population, with underlying mechanisms that remain unclear. This association might be more frequent than previously reported, indicating a need for increased clinical awareness. Understanding the interplay between FSHD and autoimmune myositis could reveal insights into the immunopathological processes of these diseases and improve diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Antonio Lauletta
- Neuromuscular Disease Centre, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy.
| | - Yves Allenbach
- Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Inserm U974, Department of Internal Medicine and Clinical Immunology, Pitié-Salpêtrière University Hospital, Paris, France
| | - Anthony Béhin
- APHP, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, Institut de Myologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Teresinha Evangelista
- Unité de Morphologie Neuromusculaire, Institut de Myologie, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - Sarah Léonard-Louis
- Service de Neuromyologie, GH Pitié-Salpêtrière, University Hospital, 75013 Paris, France
| | - Matteo Garibaldi
- Neuromuscular Disease Centre, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Olivier Benveniste
- Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Inserm U974, Department of Internal Medicine and Clinical Immunology, Pitié-Salpêtrière University Hospital, Paris, France.
| |
Collapse
|
3
|
Pacilio S, Lombardi S, Costa R, Paris F, Petrocelli G, Marrazzo P, Cenacchi G, Alviano F. Role of Perinatal Stem Cell Secretome as Potential Therapy for Muscular Dystrophies. Biomedicines 2025; 13:458. [PMID: 40002871 PMCID: PMC11852414 DOI: 10.3390/biomedicines13020458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammation mechanisms play a critical role in muscle homeostasis, and in Muscular Dystrophies (MDs), the myofiber damage triggers chronic inflammation which significantly controls the disease progression. Immunomodulatory strategies able to target inflammatory pathways and mitigate the immune-mediated damage in MDs may provide new therapeutic options. Owing to its capacity of influencing the immune response and enhancing tissue repair, stem cells' secretome has been proposed as an adjunct or standalone treatment for MDs. In this review study, we discuss the challenging points related to the inflammation condition characterizing MD pathology and provide a concise summary of the literature supporting the potential of perinatal stem cells in targeting and modulating the MD inflammation.
Collapse
Affiliation(s)
- Serafina Pacilio
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Sara Lombardi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Roberta Costa
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Francesca Paris
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Giovannamaria Petrocelli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Pasquale Marrazzo
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Giovanna Cenacchi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Francesco Alviano
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| |
Collapse
|
4
|
Leckie J, Rodriguez SH, Krahn M, Yokota T. Analysis of Exon Skipping Applicability for Dysferlinopathies. Cells 2025; 14:177. [PMID: 39936969 PMCID: PMC11817064 DOI: 10.3390/cells14030177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 02/13/2025] Open
Abstract
Exon skipping, mediated through antisense oligonucleotides (ASOs), is a promising approach to exclude pathogenic variants from the DYSF gene and treat dysferlinopathies. Understanding the applicability of various exon skipping strategies in the total patient population, an analysis not previously performed, can help guide researchers in prioritizing therapies with the broadest potential impact. Using data from the UMD-DYSF database, we evaluated all reported pathogenic variants in dysferlinopathy patients for the applicability of single- or double-exon skipping approaches to exclude the pathogenic variants while maintaining the open reading frame. A total of 61 theoretically applicable exon skipping strategies were identified, with the potential to address 90.0% of the pathogenic variants reported-44.6% through single-exon skipping and 45.3% through double-exon skipping. The most broadly applicable targets include exons 28 and 29 (9.0%), exons 27 and 28 (6.7%), and exons 50 and 51 (5.4%). While numerous theoretically applicable strategies were identified, it remains unclear if the truncated proteins produced through each exon skipping strategy will have improved functionality to alleviate patient symptoms. Further preclinical studies and clinical trials will be essential to determine the effectiveness of these therapies, potentially expanding access to disease-modifying treatments for dysferlinopathy patients.
Collapse
Affiliation(s)
- Jamie Leckie
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (J.L.); (S.H.R.)
| | - Sebastian Hernandez Rodriguez
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (J.L.); (S.H.R.)
| | - Martin Krahn
- INSERM, Marseille Medical Genetics, U1251, Aix-Marseille University, APHM, 13385 Marseille, France;
- Medical Genetics Department, Timone Hospital, APHM, 13005 Marseille, France
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G 2H7, Canada; (J.L.); (S.H.R.)
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada HM Toupin Neurological Sciences Research, Edmonton, AB T6G 2H7, Canada
| |
Collapse
|
5
|
Huang HL, Grandinetti G, Heissler SM, Chinthalapudi K. Cryo-EM structures of the membrane repair protein dysferlin. Nat Commun 2024; 15:9650. [PMID: 39511170 PMCID: PMC11544258 DOI: 10.1038/s41467-024-53773-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 10/22/2024] [Indexed: 11/15/2024] Open
Abstract
Plasma membrane repair in response to damage is essential for cell viability. The ferlin family protein dysferlin plays a key role in Ca2+-dependent membrane repair in striated muscles. Mutations in dysferlin lead to a spectrum of diseases known as dysferlinopathies. The lack of a structure of dysferlin and other ferlin family members has impeded a mechanistic understanding of membrane repair mechanisms and the development of therapies. Here, we present the cryo-EM structures of the full-length human dysferlin monomer and homodimer at 2.96 Å and 4.65 Å resolution. These structures define the architecture of dysferlin, ferlin family-specific domains, and homodimerization mechanisms essential to function. Furthermore, biophysical and cell biology studies revealed how missense mutations in dysferlin contribute to disease mechanisms. In summary, our study provides a framework for the molecular mechanisms of dysferlin and the broader ferlin family, offering a foundation for the development of therapeutic strategies aimed at treating dysferlinopathies.
Collapse
Affiliation(s)
- Hsiang-Ling Huang
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, USA
| | - Giovanna Grandinetti
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, USA
- Center for Electron Microscopy and Analysis, The Ohio State University, Columbus, USA
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, USA.
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, USA.
| |
Collapse
|
6
|
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
|
7
|
Vásquez W, Toro CA, Cardozo CP, Cea LA, Sáez JC. Pathophysiological role of connexin and pannexin hemichannels in neuromuscular disorders. J Physiol 2024. [PMID: 39173050 DOI: 10.1113/jp286173] [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: 05/01/2024] [Accepted: 07/17/2024] [Indexed: 08/24/2024] Open
Abstract
A growing body of research has provided evidence that de novo expression of connexin hemichannels and upregulation of pannexin hemichannels (Cx HCs and Panx HCs, respectively) in the cytoplasmic membrane of skeletal muscle (sarcolemma) are critical steps in the pathogenesis of muscle dysfunction of many genetic and acquired muscle diseases. This review provides an overview of the current understanding of the molecular mechanisms regulating the expression of Cx and Panx HCs in skeletal muscle, as well as their roles in both muscle physiology and pathologies. Additionally, it addresses existing gaps in knowledge and outlines future challenges in the field.
Collapse
Affiliation(s)
- Walter Vásquez
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Santiago, Chile
- Instituto de Neurociencias, Centro Interdisciplinario De Neurociencia De Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | - Carlos A Toro
- Spinal Cord Damage Research Center, James J Peters VA Medical Center, Bronx, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Christopher P Cardozo
- Spinal Cord Damage Research Center, James J Peters VA Medical Center, Bronx, NY, USA
- Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Luis A Cea
- Instituto de Ciencias Biomédicas, Facultad de Ciencias de la Salud, Universidad Autónoma de Chile, Santiago, Chile
| | - Juan C Sáez
- Instituto de Neurociencias, Centro Interdisciplinario De Neurociencia De Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| |
Collapse
|
8
|
Mahon N, Glennon JC. The Bi-directional Relationship Between Sleep and Inflammation in Muscular Dystrophies: A Narrative Review. Neurosci Biobehav Rev 2023; 150:105116. [PMID: 36870583 DOI: 10.1016/j.neubiorev.2023.105116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 01/31/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Muscular dystrophies vary in presentation and severity, but are associated with profound disability in many people. Although characterised by muscle weakness and wasting, there is also a very high prevalence of sleep problems and disorders which have significant impacts on quality of life in these individuals. There are no curative therapies for muscular dystrophies, with the only options for patients being supportive therapies to aid with symptoms. Therefore, there is an urgent need for new therapeutic targets and a greater understanding of pathogenesis. Inflammation and altered immunity are factors which have prominent roles in some muscular dystrophies and emerging roles in others such as type 1 myotonic dystrophy, signifying a link to pathogenesis. Interestingly, there is also a strong link between inflammation/immunity and sleep. In this review, we will explore this link in the context of muscular dystrophies and how it may influence potential therapeutic targets and interventions.
Collapse
Affiliation(s)
- Niamh Mahon
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Jeffrey C Glennon
- School of Medicine, University College Dublin, Dublin, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
9
|
Park J, Moon YJ, Kim DS. Miyoshi Muscular Dystrophy Type 1 with Mutated DYSF Gene Misdiagnosed as Becker Muscular Dystrophy: A Case Report and Literature Review. Genes (Basel) 2023; 14:200. [PMID: 36672942 PMCID: PMC9859596 DOI: 10.3390/genes14010200] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/28/2022] [Accepted: 01/11/2023] [Indexed: 01/13/2023] Open
Abstract
Dysferlinopathy covers a spectrum of muscle disorder categorized by two major phenotypes, namely Miyoshi muscular dystrophy type 1 (MMD1, OMIM #254130) and limb-girdle muscular dystrophy autosomal recessive 2 (LGMDR2, OMIM #253601), and two minor symptoms, including asymptomatic hyperCKemia and distal myopathy with anterior tibial onset (DMAT, OMIM #606768). We report the first Korean MMD1 misdiagnosed as Becker muscular dystrophy (BMD), which was caused by a combination of compound heterozygous c.663 + 1G > C and p.Trp992Arg of the DYSF gene. A 70-year-old male previously diagnosed with BMD was admitted for genetic counseling. Since he was clinically suspected to have dysferlinopathy but not BMD, targeted panel sequencing was performed to discover the potential hereditary cause of the suspected muscular dystrophy in the proband. Consequently, two pathogenic single nucleotide variants of the DYSF gene, c.663 + 1G > C (rs398123800) and p.Trp992Arg (rs750028300), associated with dysferlinopathy were identified. These variants were previously reported with variant allele frequencies of 0.000455 (c.663 + 1G > C) and 0.000455 (c.2974T > C; p.Trp992Arg) in the Korean population. This report emphasizes the need for common variant screening in the diagnostic algorithms of certain muscle disorders or gene panels with potential pathogenic effects and high rates of recurrent variants.
Collapse
Affiliation(s)
- Joonhong Park
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| | - Young Jae Moon
- Department of Orthopedic Surgery, Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
- Department of Biochemistry and Molecular Biology, Jeonbuk National University Medical School, Jeonju 54907, Republic of Korea
| | - Dal Sik Kim
- Department of Laboratory Medicine, Jeonbuk National University Medical School and Hospital, Jeonju 54907, Republic of Korea
- Research Institute of Clinical Medicine of Jeonbuk National University-Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|
10
|
Tanboon J, Nishino I. Autosomal Recessive Limb-Girdle Muscular Dystrophies. CURRENT CLINICAL NEUROLOGY 2023:93-121. [DOI: 10.1007/978-3-031-44009-0_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
11
|
Savarese M, Jokela M, Udd B. Distal myopathy. HANDBOOK OF CLINICAL NEUROLOGY 2023; 195:497-519. [PMID: 37562883 DOI: 10.1016/b978-0-323-98818-6.00002-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/12/2023]
Abstract
Distal myopathies are a group of genetic, primary muscle diseases. Patients develop progressive weakness and atrophy of the muscles of forearm, hands, lower leg, or feet. Currently, over 20 different forms, presenting a variable age of onset, clinical presentation, disease progression, muscle involvement, and histological findings, are known. Some of them are dominant and some recessive. Different variants in the same gene are often associated with either dominant or recessive forms, although there is a lack of a comprehensive understanding of the genotype-phenotype correlations. This chapter provides a description of the clinicopathologic and genetic aspects of distal myopathies emphasizing known etiologic and pathophysiologic mechanisms.
Collapse
Affiliation(s)
- Marco Savarese
- Folkhälsan Research Center, Helsinki, Finland; Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland
| | - Manu Jokela
- Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland; Division of Clinical Neurosciences, Department of Neurology, Turku University Hospital, Turku, Finland
| | - Bjarne Udd
- Folkhälsan Research Center, Helsinki, Finland; Department of Medical Genetics, Medicum, University of Helsinki, Helsinki, Finland; Neuromuscular Research Center, Department of Neurology, Tampere University and University Hospital, Tampere, Finland; Department of Neurology, Vaasa Central Hospital, Vaasa, Finland.
| |
Collapse
|
12
|
Wang N, Han X, Hao S, Han J, Zhou X, Sun S, Tang J, Lu Y, Wu H, Ma S, Song X, Ji G. The clinical, myopathological, and molecular characteristics of 26 Chinese patients with dysferlinopathy: a high proportion of misdiagnosis and novel variants. BMC Neurol 2022; 22:398. [PMID: 36319958 PMCID: PMC9623978 DOI: 10.1186/s12883-022-02905-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Revised: 09/20/2022] [Accepted: 09/27/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Dysferlinopathy is an autosomal recessive muscular dystrophy caused by pathogenic variants in the dysferlin (DYSF) gene. This disease shows heterogeneous clinical phenotypes and genetic characteristics. METHODS We reviewed the clinical and pathological data as well as the molecular characteristics of 26 Chinese patients with dysferlinopathy screened by immunohistochemistry staining and pathogenic variants in DYSF genes. RESULTS Among 26 patients with dysferlinopathy, 18 patients (69.2%) presented as Limb-girdle Muscular Dystrophy Type R2 (LGMD R2), 4 (15.4%) had a phenotype of Miyoshi myopathy (MM), and 4 (15.4%) presented as asymptomatic hyperCKemia. Fifteen patients (57.7%) were originally misdiagnosed as inflammatory myopathy or other diseases. Fifteen novel variants were identified among the 40 variant sites identified in this cohort. CONCLUSION Dysferlinopathy is a clinically and genetically heterogeneous group of disorders with various phenotypes, a high proportion of novel variants, and a high rate of misdiagnosis before immunohistochemistry staining and genetic analysis.
Collapse
Affiliation(s)
- Ning Wang
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Xu Han
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Shengpu Hao
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Jingzhe Han
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | | | - Shuyan Sun
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Jin Tang
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Yanpeng Lu
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Hongran Wu
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Shaojuan Ma
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Xueqin Song
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| | - Guang Ji
- grid.452702.60000 0004 1804 3009Department of Neurology, The Second Hospital of Hebei Medical University, 050000 Shijiazhuang, Hebei People’s Republic of China ,grid.256883.20000 0004 1760 8442The Key Laboratory of Neurology (Hebei Medical University), Ministry of Education, 050000 Shijiazhuang, Hebei People’s Republic of China
| |
Collapse
|
13
|
Connolly CM, Plomp L, Paik JJ, Allenbach Y. Possible future avenues for myositis therapeutics: DM, IMNM and IBM. Best Pract Res Clin Rheumatol 2022; 36:101762. [PMID: 35778272 DOI: 10.1016/j.berh.2022.101762] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Idiopathic inflammatory myopathies (IIMs) represent a heterogeneous group of systemic autoimmune diseases characterized by immune-mediated muscle injury. As insights into pathogenesis of IIM evolve, novel therapeutic strategies have become available to optimize outcomes. Herein, we summarize novel and emerging strategies in the management of dermatomyositis (DM), immunemediated necrotizing myopathy (IMNM), and inclusion body myositis (IBM).
Collapse
Affiliation(s)
- Caoilfhionn M Connolly
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lotta Plomp
- Department of Internal Medicine and Clinical Immunology, Pitié Salpêtrière Hospital, Sorbonne University, Paris, France
| | - Julie J Paik
- Division of Rheumatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| | - Yves Allenbach
- Department of Internal Medicine and Clinical Immunology, Pitié Salpêtrière Hospital, Sorbonne University, Paris, France.
| |
Collapse
|
14
|
Contreras-Cubas C, Barajas-Olmos F, Frayre-Martínez MI, Siordia-Reyes G, Guízar-Sánchez CC, García-Ortiz H, Orozco L, Baca V. Dysferlinopathy misdiagnosed with juvenile polymyositis in the pre-symptomatic stage of hyperCKemia: a case report and literature review. BMC Med Genomics 2022; 15:139. [PMID: 35725460 PMCID: PMC9208210 DOI: 10.1186/s12920-022-01284-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 06/10/2022] [Indexed: 12/05/2022] Open
Abstract
Background Dysferlinopathy encompasses a group of rare muscular dystrophies caused by recessive mutations in the DYSF gene. The phenotype ranges from asymptomatic elevated serum creatine kinase (hyperCKemia) to selective and progressive involvement of the proximal and/or distal muscles of the limbs. Bohan and Peter criteria are the most widely used for the diagnosis of polymyositis, but they have limitations and can misclassify muscular dystrophies with inflammation as polymyositis. Most dysferlinopathy patients have muscle biopsies with inflammation and thus are vulnerable to misdiagnosis with polymyositis and inappropriate treatment with steroids and immunosuppressors.
Case presentation We describe a 14 years-old male patient who was referred for assessment of asymptomatic hyperCKemia (26,372 IU/L). An X-linked dystrophinopathy initially was ruled out by direct genetic testing. Juvenile polymyositis was considered based on muscle biopsy, creatine kinase levels, and electromyography changes. Corticosteroid treatment triggered proximal lower limb muscular weakness, and no full muscular strength recovery was observed after corticosteroid withdrawal. Based on these observations, a limb-girdle muscular dystrophy (LGMD) was suspected, and LGMDR2 was confirmed by whole exome sequencing. Conclusion We report a dysferlinopathy patient who was misdiagnosed with juvenile polymyositis and explore in a literature review how common such misdiagnoses are. With diagnosis based only on routine clinicopathological examinations, distinguishing an inflammatory myopathy from dysferlinopathy is quite difficult. We suggest that before establishing a diagnosis of “definite” or “probable” juvenile polymyositis, according to Bohan and Peter or current ACR/EULAR criteria, a muscular dystrophy must first be ruled out.
Collapse
Affiliation(s)
- Cecilia Contreras-Cubas
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, SS, Mexico City, Mexico
| | - Francisco Barajas-Olmos
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, SS, Mexico City, Mexico
| | | | | | - Claudia C Guízar-Sánchez
- Department of Physical Medicine and Rehabilitation, Hospital de Pediatría, CMN Siglo XXI IMSS, Mexico City, Mexico
| | - Humberto García-Ortiz
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, SS, Mexico City, Mexico
| | - Lorena Orozco
- Immunogenomics and Metabolic Diseases Laboratory, National Institute of Genomic Medicine, SS, Mexico City, Mexico
| | - Vicente Baca
- Department of Rheumatology, Hospital de Pediatría, CMN Siglo XXI IMSS, Mexico City, Mexico.
| |
Collapse
|
15
|
Singhal R, Lukose R, Carr G, Moktar A, Gonzales-Urday AL, Rouchka EC, Vajravelu BN. Differential Expression of Long Noncoding RNAs in Murine Myoblasts After Short Hairpin RNA-Mediated Dysferlin Silencing In Vitro: Microarray Profiling. JMIR BIOINFORMATICS AND BIOTECHNOLOGY 2022; 3:e33186. [PMID: 38935964 PMCID: PMC11135227 DOI: 10.2196/33186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/02/2022] [Accepted: 05/10/2022] [Indexed: 06/29/2024]
Abstract
BACKGROUND Long noncoding RNAs (lncRNAs) are noncoding RNA transcripts greater than 200 nucleotides in length and are known to play a role in regulating the transcription of genes involved in vital cellular functions. We hypothesized the disease process in dysferlinopathy is linked to an aberrant expression of lncRNAs and messenger RNAs (mRNAs). OBJECTIVE In this study, we compared the lncRNA and mRNA expression profiles between wild-type and dysferlin-deficient murine myoblasts (C2C12 cells). METHODS LncRNA and mRNA expression profiling were performed using a microarray. Several lncRNAs with differential expression were validated using quantitative real-time polymerase chain reaction. Gene Ontology (GO) analysis was performed to understand the functional role of the differentially expressed mRNAs. Further bioinformatics analysis was used to explore the potential function, lncRNA-mRNA correlation, and potential targets of the differentially expressed lncRNAs. RESULTS We found 3195 lncRNAs and 1966 mRNAs that were differentially expressed. The chromosomal distribution of the differentially expressed lncRNAs and mRNAs was unequal, with chromosome 2 having the highest number of lncRNAs and chromosome 7 having the highest number of mRNAs that were differentially expressed. Pathway analysis of the differentially expressed genes indicated the involvement of several signaling pathways including PI3K-Akt, Hippo, and pathways regulating the pluripotency of stem cells. The differentially expressed genes were also enriched for the GO terms, developmental process and muscle system process. Network analysis identified 8 statistically significant (P<.05) network objects from the upregulated lncRNAs and 3 statistically significant network objects from the downregulated lncRNAs. CONCLUSIONS Our results thus far imply that dysferlinopathy is associated with an aberrant expression of multiple lncRNAs, many of which may have a specific function in the disease process. GO terms and network analysis suggest a muscle-specific role for these lncRNAs. To elucidate the specific roles of these abnormally expressed noncoding RNAs, further studies engineering their expression are required.
Collapse
Affiliation(s)
- Richa Singhal
- Department of Biochemistry and Molecular Genetics, KY IDeA Networks of Biomedical Research Excellence Bioinformatics Core, University of Louisville, Louisville, KY, United States
| | - Rachel Lukose
- Department of Physician Assistant Studies, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| | - Gwenyth Carr
- Department of Medical and Molecular Biology, School of Arts and Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| | - Afsoon Moktar
- Department of Physician Assistant Studies, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| | - Ana Lucia Gonzales-Urday
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| | - Eric C Rouchka
- Department of Biochemistry and Molecular Genetics, KY IDeA Networks of Biomedical Research Excellence Bioinformatics Core, University of Louisville, Louisville, KY, United States
| | - Bathri N Vajravelu
- Department of Physician Assistant Studies, Massachusetts College of Pharmacy and Health Sciences, Boston, MA, United States
| |
Collapse
|
16
|
Depuydt CE, Goosens V, Janky R, D’Hondt A, De Bleecker JL, Noppe N, Derveaux S, Thal DR, Claeys KG. Unraveling the Molecular Basis of the Dystrophic Process in Limb-Girdle Muscular Dystrophy LGMD-R12 by Differential Gene Expression Profiles in Diseased and Healthy Muscles. Cells 2022; 11:1508. [PMID: 35563815 PMCID: PMC9104122 DOI: 10.3390/cells11091508] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 04/19/2022] [Accepted: 04/29/2022] [Indexed: 11/28/2022] Open
Abstract
Limb-girdle muscular dystrophy R12 (LGMD-R12) is caused by two mutations in anoctamin-5 (ANO5). Our aim was to identify genes and pathways that underlie LGMD-R12 and explain differences in the molecular predisposition and susceptibility between three thigh muscles that are severely (semimembranosus), moderately (vastus lateralis) or mildly (rectus femoris) affected in this disease. We performed transcriptomics on these three muscles in 16 male LGMD-R12 patients and 15 age-matched male controls. Our results showed that LGMD-R12 dystrophic muscle is associated with the expression of genes indicative of fibroblast and adipocyte replacement, such as fibroadipogenic progenitors and immune cell infiltration, while muscle protein synthesis and metabolism were downregulated. Muscle degeneration was associated with an increase in genes involved in muscle injury and inflammation, and muscle repair/regeneration. Baseline differences between muscles in healthy individuals indicated that muscles that are the most affected by LGMD-R12 have the lowest expression of transcription factor networks involved in muscle (re)generation and satellite stem cell activation. Instead, they show relative high levels of fetal/embryonic myosins, all together indicating that muscles differ in their baseline regenerative potential. To conclude, we profiled the gene expression landscape in LGMD-R12, identified baseline differences in expression levels between differently affected muscles and characterized disease-associated changes.
Collapse
Affiliation(s)
- Christophe E. Depuydt
- Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, KU Leuven, and Leuven Brain Institute (LBI), Herestraat 49, 3000 Leuven, Belgium;
| | - Veerle Goosens
- Department of Radiology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium; (V.G.); (N.N.)
| | - Rekin’s Janky
- VIB Nucleomics Core, Herestraat 49, 3000 Leuven, Belgium; (R.J.); (S.D.)
| | - Ann D’Hondt
- Department of Neurology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium;
| | - Jan L. De Bleecker
- Department of Neurology, University Hospital Gent, Corneel Heymanslaan 10, 9000 Gent, Belgium;
| | - Nathalie Noppe
- Department of Radiology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium; (V.G.); (N.N.)
| | - Stefaan Derveaux
- VIB Nucleomics Core, Herestraat 49, 3000 Leuven, Belgium; (R.J.); (S.D.)
| | - Dietmar R. Thal
- Department of Pathology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium;
- Laboratory for Neuropathology, Department of Imaging and Pathology, KU Leuven, and Leuven Brain Institute (LBI), Herestraat 49, 3000 Leuven, Belgium
| | - Kristl G. Claeys
- Laboratory for Muscle Diseases and Neuropathies, Department of Neurosciences, KU Leuven, and Leuven Brain Institute (LBI), Herestraat 49, 3000 Leuven, Belgium;
- Department of Neurology, University Hospitals Leuven, Herestraat 49, 3000 Leuven, Belgium;
| |
Collapse
|
17
|
Becker N, Moore SA, Jones KA. The inflammatory pathology of dysferlinopathy is distinct from calpainopathy, Becker muscular dystrophy, and inflammatory myopathies. Acta Neuropathol Commun 2022; 10:17. [PMID: 35135626 PMCID: PMC8822795 DOI: 10.1186/s40478-022-01320-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/19/2022] [Indexed: 12/25/2022] Open
Abstract
The descriptions of muscle pathology in dysferlinopathy patients have classically included an inflammatory infiltrate that can mimic inflammatory myopathies. Based on over 20 years of institutional experience in evaluating dystrophic and inflammatory myopathy muscle biopsies at the University of Iowa, we hypothesized the inflammatory histopathology of dysferlinopathy is more similar to limb-girdle pattern muscular dystrophies such as calpainopathy and Becker muscular dystrophy, and distinct from true inflammatory myopathies. Muscle biopsies from 32 dysferlinopathy, 30 calpainopathy, 30 Becker muscular dystrophy, and 30 inflammatory myopathies (15 each of dermatomyositis and inclusion body myositis) were analyzed through digital quantitation of CD3, CD4, CD8, CD20, and PU.1 immunostaining. The expression of MHC class I and deposition of complement C5b-9 was also evaluated. Dysferlinopathy, calpainopathy, and Becker muscular dystrophy muscle biopsies had similar numbers of inflammatory cell infiltrates and significantly fewer CD3+ T-lymphocytes than dermatomyositis (p = 0.05) and inclusion body myositis (p < 0.0001) biopsies. There was no statistically significant difference in the number of PU.1+ macrophages identified in any diagnostic group. MHC class I expression was significantly lower in the limb-girdle pattern muscular dystrophies compared to the inflammatory myopathies (p < 0.0001). In contrast, complement C5b-9 deposition was similar among dysferlinopathy, dermatomyositis, and inclusion body myositis biopsies but significantly greater than calpainopathy and Becker muscular dystrophy biopsies (p = 0.05). Compared to calpainopathy, Becker muscular dystrophy, and inflammatory myopathies, the unique profile of minimal inflammatory cell infiltrates, absent to focal MHC class I, and diffuse myofiber complement C5b-9 deposition is the pathologic signature of dysferlinopathy muscle biopsies.
Collapse
|
18
|
Saidj T, Baba Amer Y, Plonquet A, Henry A, Souvannanorath S, Relaix F, Beldi-Ferchiou A, Authier FJ. Optimized Flow Cytometry Strategy for Phenotyping Intramuscular Leukocytes: Application to the Evaluation of Myopathological Processes. J Neuropathol Exp Neurol 2022; 81:193-207. [DOI: 10.1093/jnen/nlab136] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Abstract
Phenotyping intramuscular immune cells is essential for the characterization of dysimmune/inflammatory myopathies (DIM). Flow cytometry (FC) is the most reliable technique for analyzing leukocyte subpopulations and evaluating their activation levels. We developed a purely mechanical protocol for extracting cells from muscle tissue allowing us to preserve cell surface epitopes and determined its applicability to experimental pathology in mice and myopathological diagnosis in human. Skeletal muscle regeneration in mice was associated with a transient enrichment of macrophages (CD11bhighGr-1+), myeloid dendritic cells (CD3−C8+CD11bhigh), CD8+ T cells (CD3+C8+), and NK cells (CD3− CD11bhighNKp46+). In murine models of inherited muscle dystrophies, leukocytes represented 23%–84% of intramuscular mononuclear cells, with a percentage of CD8+ T cells (4%–17%) mirroring that of all CD45+ cells, while MDCs remained a minority. In human 16 samples (DIM: n = 9; nonimmune conditions: n = 7), DIM was associated with intramuscular recruitment of CD8+ T cells, but not CD4+ T cells and NK cells. FC allowed concomitant quantification of HLA-DR, CD25, CD38, and CD57 activation/differentiation biomarkers and showed increased activation levels of CD4+ and CD8+ T cells in DIM. In conclusion, FC is an appropriate method for quantifying intramuscular leukocyte subpopulations and analyzing their activation states.
Collapse
Affiliation(s)
- Tassadit Saidj
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
| | - Yasmine Baba Amer
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
| | - Anne Plonquet
- AP-HP, Hôpitaux Universitaires Henri Mondor, Laboratoire d'immunologie Biologique, Créteil, France
| | - Adeline Henry
- Université Paris Est Créteil, INSERM, IMRB, Plateforme de Cytométrie en flux, Créteil, France
| | - Sarah Souvannanorath
- Département de Pathologie, APHP, Hôpitaux Universitaires Henri Mondor, Centre de Référence des Maladies Rares Neuromusculaire Nord/Est/Ile-de-France, ERN Euro-NMD, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
| | - Frederic Relaix
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
| | - Asma Beldi-Ferchiou
- AP-HP, Hôpitaux Universitaires Henri Mondor, Laboratoire d'immunologie Biologique, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Equipe Cohen, Créteil, France
| | - François Jérôme Authier
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
- Département de Pathologie, APHP, Hôpitaux Universitaires Henri Mondor, Centre de Référence des Maladies Rares Neuromusculaire Nord/Est/Ile-de-France, ERN Euro-NMD, Créteil, France
- Université Paris Est Créteil, INSERM, IMRB, Equipe Relaix, Creteil, France
| |
Collapse
|
19
|
Bittel DC, Sreetama SC, Chandra G, Ziegler R, Nagaraju K, Van der Meulen JH, Jaiswal JK. Secreted acid sphingomyelinase as a potential gene therapy for limb girdle muscular dystrophy 2B. J Clin Invest 2022; 132:e141295. [PMID: 34981776 PMCID: PMC8718136 DOI: 10.1172/jci141295] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 11/05/2021] [Indexed: 12/14/2022] Open
Abstract
Efficient sarcolemmal repair is required for muscle cell survival, with deficits in this process leading to muscle degeneration. Lack of the sarcolemmal protein dysferlin impairs sarcolemmal repair by reducing secretion of the enzyme acid sphingomyelinase (ASM), and causes limb girdle muscular dystrophy 2B (LGMD2B). The large size of the dysferlin gene poses a challenge for LGMD2B gene therapy efforts aimed at restoring dysferlin expression in skeletal muscle fibers. Here, we present an alternative gene therapy approach targeting reduced ASM secretion, the consequence of dysferlin deficit. We showed that the bulk endocytic ability is compromised in LGMD2B patient cells, which was addressed by extracellularly treating cells with ASM. Expression of secreted human ASM (hASM) using a liver-specific adeno-associated virus (AAV) vector restored membrane repair capacity of patient cells to healthy levels. A single in vivo dose of hASM-AAV in the LGMD2B mouse model restored myofiber repair capacity, enabling efficient recovery of myofibers from focal or lengthening contraction-induced injury. hASM-AAV treatment was safe, attenuated fibro-fatty muscle degeneration, increased myofiber size, and restored muscle strength, similar to dysferlin gene therapy. These findings elucidate the role of ASM in dysferlin-mediated plasma membrane repair and to our knowledge offer the first non-muscle-targeted gene therapy for LGMD2B.
Collapse
Affiliation(s)
- Daniel C. Bittel
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Sen Chandra Sreetama
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Goutam Chandra
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
| | - Robin Ziegler
- Rare and Neurologic Diseases Research, Sanofi, Framingham, Massachusetts, USA
| | - Kanneboyina Nagaraju
- School of Pharmacy and Pharmaceutical Sciences, SUNY Binghamton University, Binghamton, New York, USA
| | | | - Jyoti K. Jaiswal
- Center for Genetic Medicine Research, Children’s National Hospital, Washington, DC, USA
- Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| |
Collapse
|
20
|
Therapeutic Benefit of Galectin-1: Beyond Membrane Repair, a Multifaceted Approach to LGMD2B. Cells 2021; 10:cells10113210. [PMID: 34831431 PMCID: PMC8621416 DOI: 10.3390/cells10113210] [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: 09/30/2021] [Revised: 11/11/2021] [Accepted: 11/12/2021] [Indexed: 11/21/2022] Open
Abstract
Two of the main pathologies characterizing dysferlinopathies are disrupted muscle membrane repair and chronic inflammation, which lead to symptoms of muscle weakness and wasting. Here, we used recombinant human Galectin-1 (rHsGal-1) as a therapeutic for LGMD2B mouse and human models. Various redox and multimerization states of Gal-1 show that rHsGal-1 is the most effective form in both increasing muscle repair and decreasing inflammation, due to its monomer-dimer equilibrium. Dose-response testing shows an effective 25-fold safety profile between 0.54 and 13.5 mg/kg rHsGal-1 in Bla/J mice. Mice treated weekly with rHsGal-1 showed downregulation of canonical NF-κB inflammation markers, decreased muscle fat deposition, upregulated anti-inflammatory cytokines, increased membrane repair, and increased functional movement compared to non-treated mice. Gal-1 treatment also resulted in a positive self-upregulation loop of increased endogenous Gal-1 expression independent of NF-κB activation. A similar reduction in disease pathologies in patient-derived human cells demonstrates the therapeutic potential of Gal-1 in LGMD2B patients.
Collapse
|
21
|
Holm-Yildiz S, Witting N, de Stricker Borch J, Kass K, Khawajazada T, Krag T, Vissing J. Muscle biopsy and MRI findings in ANO5-related myopathy. Muscle Nerve 2021; 64:743-748. [PMID: 34550615 DOI: 10.1002/mus.27419] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 09/06/2021] [Accepted: 09/18/2021] [Indexed: 12/31/2022]
Abstract
INTRODUCTION/AIMS Mutations in the anoctamin 5 (ANO5) gene are a common cause of muscular dystrophy. We aimed to investigate whether inflammatory changes in muscle are present in patients with ANO5 myopathy when assessed by muscle biopsy and muscle magnetic resonance imaging (MRI). METHODS Adults with pathogenic variations in ANO5 known to cause muscular dystrophy were included in our study. Muscle biopsies of pelvic and lower extremity muscles were reviewed retrospectively. Muscle MR short-tau inversion recovery (STIR) images of a subset of these patients were obtained prospectively. RESULTS Muscle biopsies from 24 patients were reviewed. MR STIR images were performed in 17 of these patients. We found inflammatory changes in muscle biopsies of three patients and MRI revealed hyperintense signals on STIR images in 14 of 17 patients. DISCUSSION In this study, we found that muscle edema is very common in patients with ANO5 myopathy and that some patients have inflammatory changes in muscle biopsies. Further studies are needed to determine whether the STIR+ lesions reflect inflammation.
Collapse
Affiliation(s)
- Sonja Holm-Yildiz
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Nanna Witting
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Josefine de Stricker Borch
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Konni Kass
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Tahmina Khawajazada
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Thomas Krag
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - John Vissing
- Copenhagen Neuromuscular Center, Department of Neurology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
22
|
Diagnostic muscle biopsies in the era of genetics: the added value of myopathology in a selection of limb-girdle muscular dystrophy patients. Acta Neurol Belg 2021; 121:1019-1033. [PMID: 33400223 DOI: 10.1007/s13760-020-01559-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2020] [Accepted: 11/19/2020] [Indexed: 10/22/2022]
Abstract
In the second most common dystrophy associated with predominant pelvic and shoulder girdle muscle weakness termed Limb-Girdle Muscular Dystrophy (LGMD), genetic complexity, large phenotypic variability, and clinical overlap can result in extensive diagnostic delays in certain individuals. In view of the large strides genetics has taken in this day and age, we address the question if muscle biopsies can still provide diagnostic evidence of substance for these patients. We reviewed and reanalyzed muscle biopsy characteristics in a cohort of LGMD patient pairs in which gene variants were picked up in CAPN3, FKRP, TTN, and ANO5, using histochemical-immunohistochemical-and immunofluorescent staining, and western blotting. We found that not the nature and severity of inflammatory changes, but the changed properties of the dystrophin complex were the most valuable assets to differentiate LGMD from myositis. Proteomic evaluation brought both primary and secondary deficiencies to light, which could be equally revealing for diagnosis. Though a muscle biopsy might, at present, not always be strictly necessary anymore, it still represents an irrefutable asset when the genetic diagnosis is complicated.
Collapse
|
23
|
Nicolau S, Milone M, Liewluck T. Guidelines for genetic testing of muscle and neuromuscular junction disorders. Muscle Nerve 2021; 64:255-269. [PMID: 34133031 DOI: 10.1002/mus.27337] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 12/24/2022]
Abstract
Despite recent advances in the understanding of inherited muscle and neuromuscular junction diseases, as well as the advent of a wide range of genetic tests, patients continue to face delays in diagnosis of sometimes treatable disorders. These guidelines outline an approach to genetic testing in such disorders. Initially, a patient's phenotype is evaluated to identify myopathies requiring directed testing, including myotonic dystrophies, facioscapulohumeral muscular dystrophy, oculopharyngeal muscular dystrophy, mitochondrial myopathies, dystrophinopathies, and oculopharyngodistal myopathy. Initial investigation in the remaining patients is generally a comprehensive gene panel by next-generation sequencing. Broad panels have a higher diagnostic yield and can be cost-effective. Due to extensive phenotypic overlap and treatment implications, genes responsible for congenital myasthenic syndromes should be included when evaluating myopathy patients. For patients whose initial genetic testing is negative or inconclusive, phenotypic re-evaluation is warranted, along with consideration of genes and variants not included initially, as well as their acquired mimickers.
Collapse
Affiliation(s)
- Stefan Nicolau
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | - Teerin Liewluck
- Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
24
|
Reactive Changes in Elements of Stromal-Vascular Differons of Dysferlin-Deficient Skeletal Muscles after Procaine Injection. Bull Exp Biol Med 2021; 170:677-681. [PMID: 33788118 DOI: 10.1007/s10517-021-05131-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Indexed: 10/21/2022]
Abstract
The study assessed reactivity of stromal-vascular skeletal muscle differons to acute chemical injury. Dysferlin-deficient Bla/J mice and the wild-type С57BL/6 mice were intramuscularly injected with 100 μl of 0.5% procaine solution. The middle segment of gastrocnemius muscle was taken on postsurgery days 2, 4, 10, and 14 for routine histological examination. To evaluate proliferation and vascularization, the paraffin sections were stained immunohistochemically with antibodies to α-smooth muscle actin and Ki-67. The connective tissue was stained according to Mallory. The study revealed diminished proliferative activity of stromal-vascular differons and decreased vascular density in muscles of Bla/J mice. Thus, mutations in the DYSF gene coding dysferlin down-regulate the reparation processes in all differons of skeletal muscle.
Collapse
|
25
|
Dexamethasone accelerates muscle regeneration by modulating kinesin-1-mediated focal adhesion signals. Cell Death Discov 2021; 7:35. [PMID: 33597503 PMCID: PMC7889929 DOI: 10.1038/s41420-021-00412-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 10/27/2020] [Accepted: 10/30/2020] [Indexed: 01/11/2023] Open
Abstract
During differentiation, skeletal muscle develops mature multinucleated muscle fibers, which could contract to exert force on a substrate. Muscle dysfunction occurs progressively in patients with muscular dystrophy, leading to a loss of the ability to walk and eventually to death. The synthetic glucocorticoid dexamethasone (Dex) has been used therapeutically to treat muscular dystrophy by an inhibition of inflammation, followed by slowing muscle degeneration and stabilizing muscle strength. Here, in mice with muscle injury, we found that Dex significantly promotes muscle regeneration via promoting kinesin-1 motor activity. Nevertheless, how Dex promotes myogenesis through kinesin-1 motors remains unclear. We found that Dex directly increases kinesin-1 motor activity, which is required for the expression of a myogenic marker (muscle myosin heavy chain 1/2), and also for the process of myoblast fusion and the formation of polarized myotubes. Upon differentiation, kinesin-1 mediates the recruitment of integrin β1 onto microtubules allowing delivery of the protein into focal adhesions. Integrin β1-mediated focal adhesion signaling then guides myoblast fusion towards a polarized morphology. By imposing geometric constrains via micropatterns, we have proved that cell adhesion is able to rescue the defects caused by kinesin-1 inhibition during the process of myogenesis. These discoveries reveal a mechanism by which Dex is able to promote myogenesis, and lead us towards approaches that are more efficient in improving skeletal muscle regeneration.
Collapse
|
26
|
Liang WC, Jong YJ, Wang CH, Wang CH, Tian X, Chen WZ, Kan TM, Minami N, Nishino I, Wong LJC. Clinical, pathological, imaging, and genetic characterization in a Taiwanese cohort with limb-girdle muscular dystrophy. Orphanet J Rare Dis 2020; 15:160. [PMID: 32576226 PMCID: PMC7310488 DOI: 10.1186/s13023-020-01445-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 06/16/2020] [Indexed: 12/14/2022] Open
Abstract
Background Limb-girdle muscular dystrophy (LGMD) is a genetically heterogeneous, hereditary disease characterized by limb-girdle weakness and histologically dystrophic changes. The prevalence of each subtype of LGMD varies among different ethnic populations. This study for the first time analyzed the phenotypes and genotypes in Taiwanese patients with LGMD in a referral center for neuromuscular diseases (NMDs). Results We enrolled 102 patients clinically suspected of having LGMD who underwent muscle biopsy with subsequent genetic analysis in the previous 10 years. On the basis of different pathological categories, we performed sequencing of target genes or panel for NMDs and then identified patients with type 1B, 1E, 2A, 2B, 2D, 2I, 2G, 2 N, and 2Q. The 1B patients with LMNA mutation presented with mild limb-girdle weakness but no conduction defect at the time. All 1E patients with DES mutation exhibited predominantly proximal weakness along with distal weakness. In our cohort, 2B and 2I were the most frequent forms of LGMD; several common or founder mutations were identified, including c.1097_1099delACA (p.Asn366del) in DES, homozygous c.101G > T (p.Arg34Leu) in SGCA, homozygous c.26_33dup (p.Glu12Argfs*20) in TCAP, c.545A > G (p.Tyr182Cys), and c.948delC (p.Cys317Alafs*111) in FKRP. Clinically, the prevalence of dilated cardiomyopathy in our patients with LGMD2I aged > 18 years was 100%, much higher than that in European cohorts. The only patient with LGMD2Q with PLEC mutation did not exhibit skin lesions or gastrointestinal abnormalities but had mild facial weakness. Muscle imaging of LGMD1E and 2G revealed a more uniform involvement than did other LGMD types. Conclusion Our study revealed that detailed clinical manifestation together with muscle pathology and imaging remain critical in guiding further molecular analyses and are crucial for establishing genotype–phenotype correlations. We also determined the common mutations and prevalence for different subtypes of LGMD in our cohort, which could be useful when providing specific care and personalized therapy to patients with LGMD.
Collapse
Affiliation(s)
- Wen-Chen Liang
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Translational Research Center of Neuromuscular Diseases, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan.,Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuh-Jyh Jong
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Translational Research Center of Neuromuscular Diseases, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Department of Laboratory Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan. .,Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | - Chien-Hua Wang
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chen-Hua Wang
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Xia Tian
- Baylor Genetics, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Wan-Zi Chen
- Department of Pathology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Tzu-Min Kan
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Narihiro Minami
- Department of Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Ichizo Nishino
- Department of Neuromuscular Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan.,Department of Genome Medicine Development, Medical Genome Center, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Lee-Jun C Wong
- Baylor Genetics, Houston, TX, USA.,Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
27
|
Greenberg SA. Inclusion body myositis: clinical features and pathogenesis. Nat Rev Rheumatol 2020; 15:257-272. [PMID: 30837708 DOI: 10.1038/s41584-019-0186-x] [Citation(s) in RCA: 165] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Inclusion body myositis (IBM) is often viewed as an enigmatic disease with uncertain pathogenic mechanisms and confusion around diagnosis, classification and prospects for treatment. Its clinical features (finger flexor and quadriceps weakness) and pathological features (invasion of myofibres by cytotoxic T cells) are unique among muscle diseases. Although IBM T cell autoimmunity has long been recognized, enormous attention has been focused for decades on several biomarkers of myofibre protein aggregates, which are present in <1% of myofibres in patients with IBM. This focus has given rise, together with the relative treatment refractoriness of IBM, to a competing view that IBM is not an autoimmune disease. Findings from the past decade that implicate autoimmunity in IBM include the identification of a circulating autoantibody (anti-cN1A); the absence of any statistically significant genetic risk factor other than the common autoimmune disease 8.1 MHC haplotype in whole-genome sequencing studies; the presence of a marked cytotoxic T cell signature in gene expression studies; and the identification in muscle and blood of large populations of clonal highly differentiated cytotoxic CD8+ T cells that are resistant to many immunotherapies. Mounting evidence that IBM is an autoimmune T cell-mediated disease provides hope that future therapies directed towards depleting these cells could be effective.
Collapse
Affiliation(s)
- Steven A Greenberg
- Department of Neurology, Brigham and Women's Hospital, Boston, MA, USA. .,Children's Hospital Computational Health Informatics Program, Boston Children's Hospital, Boston, MA, USA. .,Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
28
|
Fibroadipogenic progenitors are responsible for muscle loss in limb girdle muscular dystrophy 2B. Nat Commun 2019; 10:2430. [PMID: 31160583 PMCID: PMC6547715 DOI: 10.1038/s41467-019-10438-z] [Citation(s) in RCA: 102] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2017] [Accepted: 05/10/2019] [Indexed: 12/30/2022] Open
Abstract
Muscle loss due to fibrotic or adipogenic replacement of myofibers is common in muscle diseases and muscle-resident fibro/adipogenic precursors (FAPs) are implicated in this process. While FAP-mediated muscle fibrosis is widely studied in muscle diseases, the role of FAPs in adipogenic muscle loss is not well understood. Adipogenic muscle loss is a feature of limb girdle muscular dystrophy 2B (LGMD2B) - a disease caused by mutations in dysferlin. Here we show that FAPs cause the adipogenic loss of dysferlin deficient muscle. Progressive accumulation of Annexin A2 (AnxA2) in the myofiber matrix causes FAP differentiation into adipocytes. Lack of AnxA2 prevents FAP adipogenesis, protecting against adipogenic loss of dysferlinopathic muscle while exogenous AnxA2 enhances muscle loss. Pharmacological inhibition of FAP adipogenesis arrests adipogenic replacement and degeneration of dysferlin-deficient muscle. These results demonstrate the pathogenic role of FAPs in LGMD2B and establish these cells as therapeutic targets to ameliorate muscle loss in patients.
Collapse
|
29
|
Lin HT, Liu X, Zhang W, Liu J, Zuo YH, Xiao JX, Zhu Y, Yuan Y, Wang ZX. Muscle Magnetic Resonance Imaging in Patients with Various Clinical Subtypes of LMNA-Related Muscular Dystrophy. Chin Med J (Engl) 2018; 131:1472-1479. [PMID: 29893365 PMCID: PMC6006825 DOI: 10.4103/0366-6999.233957] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Background: LMNA-related muscular dystrophy can manifest in a wide variety of disorders, including Emery-Dreifuss muscular dystrophy (EDMD), limb-girdle muscular dystrophy (LGMD), and LMNA-associated congenital muscular dystrophy (L-CMD). Muscle magnetic resonance imaging (MRI) has become a useful tool in the diagnostic workup of patients with muscle dystrophies. This study aimed to investigate whether there is a consistent pattern of MRI changes in patients with LMNA mutations in various muscle subtypes. Methods: Twenty-two patients with LMNA-related muscular dystrophies were enrolled in this study. MRI of the thigh and/or calf muscles was performed in them. The muscle MRI features of the three subtypes were compared by the Mann-Whitney U-test. The relationship between the clinical and MRI findings was also investigated by Spearman's rank analyses. Results: The present study included five EDMD, nine LGMD, and eight L-CMD patients. The thigh muscle MRI revealed that the fatty infiltration of the adductor magnus, semimembranosus, long and short heads of the biceps femoris, and vasti muscles, with relative sparing of the rectus femoris, was the predominant change observed in the EDMD, LGMD, and advanced-stage L-CMD phenotypes, although the involvement of the vasti muscles was not prominent in the early stage of L-CMD. At the level of the calf, six patients (one EDMD, four LGMD, and one L-CMD) also showed a similar pattern, in which the soleus and the medial and lateral gastrocnemius muscles were most frequently observed to have fatty infiltration. The fatty infiltration severity demonstrated higher scores associated with disease progression, with a corresponding rate of 1.483 + 0.075 × disease duration (X) (r = 0.444, P = 0.026). It was noteworthy that in six L-CMD patients with massive inflammatory cell infiltration in muscle pathology, no remarkable edema-like signals were observed in muscle MRI. Conclusions: EDMD, LGMD and advanced-staged L-CMD subtypes showed similar pattern of muscle MRI changes, while early-staged L-CMD showed somewhat different changes. Muscle MRI of L-CMD with a muscular dystrophy pattern in MRI provided important clues for differentiating it from childhood inflammatory myopathy. The fatty infiltration score could be used as a reliable biomarker for outcome measure of disease progression.
Collapse
Affiliation(s)
- Hui-Ting Lin
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Xiao Liu
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Wei Zhang
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Jing Liu
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Yue-Huan Zuo
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Jiang-Xi Xiao
- Department of Radiology, Peking University First Hospital, Beijing 100034, China
| | - Ying Zhu
- Department of Radiology, Peking University First Hospital, Beijing 100034, China
| | - Yun Yuan
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| | - Zhao-Xia Wang
- Department of Neurology, Peking University First Hospital, Beijing 100034, China
| |
Collapse
|
30
|
Abstract
The immune response to acute muscle damage is important for normal repair. However, in chronic diseases such as many muscular dystrophies, the immune response can amplify pathology and play a major role in determining disease severity. Muscular dystrophies are inheritable diseases that vary tremendously in severity, but share the progressive loss of muscle mass and function that can be debilitating and lethal. Mutations in diverse genes cause muscular dystrophy, including genes that encode proteins that maintain membrane strength, participate in membrane repair, or are components of the extracellular matrix or the nuclear envelope. In this article, we explore the hypothesis that an important feature of many muscular dystrophies is an immune response adapted to acute, infrequent muscle damage that is misapplied in the context of chronic injury. We discuss the involvement of the immune system in the most common muscular dystrophy, Duchenne muscular dystrophy, and show that the immune system influences muscle death and fibrosis as disease progresses. We then present information on immune cell function in other muscular dystrophies and show that for many muscular dystrophies, release of cytosolic proteins into the extracellular space may provide an initial signal, leading to an immune response that is typically dominated by macrophages, neutrophils, helper T-lymphocytes, and cytotoxic T-lymphocytes. Although those features are similar in many muscular dystrophies, each muscular dystrophy shows distinguishing features in the magnitude and type of inflammatory response. These differences indicate that there are disease-specific immunomodulatory molecules that determine response to muscle cell damage caused by diverse genetic mutations. © 2018 American Physiological Society. Compr Physiol 8:1313-1356, 2018.
Collapse
Affiliation(s)
- James G. Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Steven S. Welc
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| |
Collapse
|
31
|
Sreetama SC, Chandra G, Van der Meulen JH, Ahmad MM, Suzuki P, Bhuvanendran S, Nagaraju K, Hoffman EP, Jaiswal JK. Membrane Stabilization by Modified Steroid Offers a Potential Therapy for Muscular Dystrophy Due to Dysferlin Deficit. Mol Ther 2018; 26:2231-2242. [PMID: 30166241 PMCID: PMC6127637 DOI: 10.1016/j.ymthe.2018.07.021] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Revised: 07/15/2018] [Accepted: 07/24/2018] [Indexed: 11/16/2022] Open
Abstract
Mutations of the DYSF gene leading to reduced dysferlin protein level causes limb girdle muscular dystrophy type 2B (LGMD2B). Dysferlin facilitates sarcolemmal membrane repair in healthy myofibers, thus its deficit compromises myofiber repair and leads to chronic muscle inflammation. An experimental therapeutic approach for LGMD2B is to protect damage or improve repair of myofiber sarcolemma. Here, we compared the effects of prednisolone and vamorolone (a dissociative steroid; VBP15) on dysferlin-deficient myofiber repair. Vamorolone, but not prednisolone, stabilized dysferlin-deficient muscle cell membrane and improved repair of dysferlin-deficient mouse (B6A/J) myofibers injured by focal sarcolemmal damage, eccentric contraction-induced injury or injury due to spontaneous in vivo activity. Vamorolone decreased sarcolemmal lipid mobility, increased muscle strength, and decreased late-stage myofiber loss due to adipogenic infiltration. In contrast, the conventional glucocorticoid prednisolone failed to stabilize dysferlin deficient muscle cell membrane or improve repair of dysferlinopathic patient myoblasts and mouse myofibers. Instead, prednisolone treatment increased muscle weakness and myofiber atrophy in B6A/J mice—findings that correlate with reports of prednisolone worsening symptoms of LGMD2B patients. Our findings showing improved cellular and pre-clinical efficacy of vamorolone compared to prednisolone and better safety profile of vamorolone indicates the suitability of vamorolone for clinical trials in LGMD2B.
Collapse
Affiliation(s)
- Sen Chandra Sreetama
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Goutam Chandra
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Jack H Van der Meulen
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Mohammad Mahad Ahmad
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Peter Suzuki
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Shivaprasad Bhuvanendran
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA
| | - Kanneboyina Nagaraju
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA
| | - Eric P Hoffman
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, Binghamton University, Binghamton, NY 13902, USA
| | - Jyoti K Jaiswal
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC 20010, USA; Department of Genomics and Precision Medicine, George Washington University School of Medicine and Health Sciences, Washington, DC 20010, USA.
| |
Collapse
|
32
|
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
|
33
|
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
|
34
|
Tang J, Song X, Ji G, Wu H, Sun S, Lu S, Li Y, Zhang C, Zhang H. A novel mutation in the DYSF gene in a patient with a presumed inflammatory myopathy. Neuropathology 2018; 38:433-437. [PMID: 29799141 DOI: 10.1111/neup.12474] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 04/05/2018] [Accepted: 04/09/2018] [Indexed: 01/30/2023]
Abstract
Dysferlinopathy, a progressive muscular dystrophy, results from mutations in the Dysferlin gene (DYSF, MIM*603009). Traditional diagnosis relies on the reduction or absence of dysferlin. However, altered dysferlin has been observed in other myopathies, leading to a precise diagnosis through molecular genetics. In this study, we report a patient who was previously misdiagnosed as inflammatory myopathy based on routine clinicopathological examinations alone. However, muscle biopsy specimens were analyzed further by immunohistochemistry of muscular dystrophy-related proteins, and gene-targeted next generation sequencing (NGS) was used to correctly identify muscular dystrophy. DNA was sequenced with NGS and the detected mutation was verified by Sanger sequencing. Our targeted NGS found a novel missense mutation (c.5392G > A) in the DYSF gene, allowing correct diagnosis of LGMD2B in our patient. We discovered of a novel missense mutation in the DYSF gene and have broadened the DYSF mutation spectrum, which may be correlated in patients with presumed dysferlinopathy, especially when lymphocytic infiltration is observed.
Collapse
Affiliation(s)
- Jin Tang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiahzuang, China
| | - Xueqin Song
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiahzuang, China
| | - Guang Ji
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiahzuang, China
| | - Hongran Wu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiahzuang, China
| | - Shuyan Sun
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiahzuang, China
| | - Shan Lu
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiahzuang, China
| | - Yuan Li
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiahzuang, China
| | - Chi Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiahzuang, China
| | - Huiqing Zhang
- Department of Neurology, The Second Hospital of Hebei Medical University, Shijiahzuang, China
| |
Collapse
|
35
|
Xu C, Chen J, Zhang Y, Li J. Limb-girdle muscular dystrophy type 2B misdiagnosed as polymyositis at the early stage: Case report and literature review. Medicine (Baltimore) 2018; 97:e10539. [PMID: 29794729 PMCID: PMC6392577 DOI: 10.1097/md.0000000000010539] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
RATIONALE Dysferlin myopathy is an autosomal recessive hereditary muscular dystrophy due to deficiency of dysferlin caused by alteration of the DYSF gene; Limb-girdle muscular dystrophy type 2B (LGMD2B) is the most common in Its clinical phenotypes. However, LGMD2B is rarely seen in clinical cases and may initially present as weakness of proximalpelvis muscles and muscles in the posterior compartments of thighs,which will then cause difficulty in running and limping during walking. Laboratory tests at an early stage of the disease often indicate an increased level of serum creatine kinase (CK). Moreover, polymyositis (PM) is manifested as symmetrical proximal muscle weakness of the four limbs, accompanied by an increased level of serum CK. Thus, both are very difficult to identify in clinical practice. PATIENT CONCERNS A 25-year-old woman was admitted to our department as the limb weakness progressively worsened. She began to experience proximal muscle weakness of both lower limbs without obvious inducement, which markedly increased when she climbed the stairs or stood up after squatting. Then her symptoms worsened, with difficulty in proximal and distal lifting of the lower extremities. DIAGNOSES Through combined immunohistochemistry and Western-blot analysis, The patient was diagnosed with LGMD2B. INTERVENTIONS There were symptomatic treatments such as coenzyme Q10. OUTCOMES After symptomatic treatments, the patient's symptoms were obviously relieved, and the CK level decreased. LESSONS Through this case, we found that combined application of immunohistochemistry and Western-blot analysis is helpful in early diagnosis of LGMD2B, and a new site of frame-shift mutation in the patient's DYSF gene was found.
Collapse
|
36
|
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 2018; 364:409-419. [PMID: 29284661 PMCID: PMC5801553 DOI: 10.1124/jpet.117.244244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [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
|
37
|
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
|
38
|
Angelini C, Fanin M. Limb girdle muscular dystrophies: clinical-genetical diagnostic update and prospects for therapy. Expert Opin Orphan Drugs 2017. [DOI: 10.1080/21678707.2017.1367283] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Corrado Angelini
- Department of Neurodegenerative Disorders, Neuromuscular Center, San Camillo Hospital IRCCS, Venice, Italy
| | - Marina Fanin
- Department of Neurosciences, University of Padova, Padova, Italy
| |
Collapse
|
39
|
Baek JH, Many GM, Evesson FJ, Kelley VR. Dysferlinopathy Promotes an Intramuscle Expansion of Macrophages with a Cyto-Destructive Phenotype. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:1245-1257. [PMID: 28412297 DOI: 10.1016/j.ajpath.2017.02.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 02/14/2017] [Indexed: 01/05/2023]
Abstract
Dysferlinopathies are a group of muscular dystrophies resulting from a genetic deficiency in Dysf. Macrophages, highly plastic cells that mediate tissue repair and destruction, are prominent within dystrophic skeletal muscles of dysferlinopathy patients. We hypothesized that Dysf-deficient muscle promotes recruitment, proliferation, and skewing of macrophages toward a cyto-destructive phenotype in dysferlinopathy. To track macrophage dynamics in dysferlinopathy, we adoptively transferred enhanced green fluorescent protein-labeled monocytes into Dysf-deficient BLA/J mice with age-related (2 to 10 months) muscle disease and Dysf-intact (C57BL/6 [B6]) mice. We detected an age- and disease-related increase in monocyte recruitment into Dysf-deficient muscles. Moreover, macrophages recruited into muscle proliferated locally and were skewed toward a cyto-destructive phenotype. By comparing Dysf-deficient and -intact monocytes, our data showed that Dysf in muscle, but not in macrophages, mediate intramuscle macrophage recruitment and proliferation. To further elucidate macrophage mechanisms related to dysferlinopathy, we investigated in vitro macrophage-myogenic cell interactions and found that Dysf-deficient muscle i) promotes macrophage proliferation, ii) skews macrophages toward a cyto-destructive phenotype, and iii) is more vulnerable to macrophage-mediated apoptosis. Taken together, our data suggest that the loss of Dysf expression in muscle, not macrophages, promotes the intramuscle expansion of cyto-destructive macrophages likely to contribute to dysferlinopathy. Identifying pathways within the Dysf-deficient muscle milieu that regulate cyto-destructive macrophages will potentially uncover therapeutic strategies for dysferlinopathies.
Collapse
Affiliation(s)
- Jea-Hyun Baek
- Laboratory of Molecular Autoimmune Disease, Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Gina M Many
- Laboratory of Molecular Autoimmune Disease, Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Frances J Evesson
- Department of Cell Biology, Harvard Medical School and Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, Massachusetts
| | - Vicki R Kelley
- Laboratory of Molecular Autoimmune Disease, Renal Division, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts.
| |
Collapse
|
40
|
Scalco RS, Lorenzoni PJ, Lynch DS, Martins WA, Jungbluth H, Quinlivan R, Becker J, Houlden H. Polymyositis without Beneficial Response to Steroid Therapy: Should Miyoshi Myopathy be a Differential Diagnosis? AMERICAN JOURNAL OF CASE REPORTS 2017; 18:17-21. [PMID: 28053302 PMCID: PMC5228759 DOI: 10.12659/ajcr.900970] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Patient: Male, 16 Final Diagnosis: Miyoshi myopathy Symptoms: HyperCKemia • myalgia • weakness Medication: — Clinical Procedure: — Specialty: Neurology
Collapse
Affiliation(s)
- Renata Siciliani Scalco
- MRC Centre for Neuromuscular Diseases and Division of Molecular Neuroscience, University College London (UCL) Institute of Neurology, London, United Kingdom.,Department of Neurology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil.,Capes Foundation, Ministry of Education, Brasilia, Brazil
| | - Paulo José Lorenzoni
- Service of Neuromuscular Disorders, Division of Neurology, Department of Internal Medicine, Universidade Federal do Paraná (UFPR), Curitiba, Brazil
| | - David S Lynch
- MRC Centre for Neuromuscular Diseases and Division of Molecular Neuroscience, University College London (UCL) Institute of Neurology, London, United Kingdom
| | - William Alves Martins
- Department of Neurology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Heinz Jungbluth
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London (KCL), London, United Kingdom.,Department of Paediatric Neurology, Evelina Children's Hospital, Guy's and St Thomas NHS Foundation Trust, London, United Kingdom.,Randall Division for Cell and Molecular Biophysics, Muscle Signalling Section, King's College London, London, United Kingdom
| | - Ros Quinlivan
- MRC Centre for Neuromuscular Diseases and Division of Molecular Neuroscience, University College London (UCL) Institute of Neurology, London, United Kingdom
| | - Jefferson Becker
- Department of Neurology, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Henry Houlden
- MRC Centre for Neuromuscular Diseases and Division of Molecular Neuroscience, University College London (UCL) Institute of Neurology, London, United Kingdom
| |
Collapse
|
41
|
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: 67] [Impact Index Per Article: 7.4] [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
|
42
|
Vincent AE, Rosa HS, Alston CL, Grady JP, Rygiel KA, Rocha MC, Barresi R, Taylor RW, Turnbull DM. Dysferlin mutations and mitochondrial dysfunction. Neuromuscul Disord 2016; 26:782-788. [PMID: 27666772 PMCID: PMC5091283 DOI: 10.1016/j.nmd.2016.08.008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 08/03/2016] [Accepted: 08/15/2016] [Indexed: 12/22/2022]
Abstract
Dysferlinopathies are caused by mutations in the DYSF gene and patients may present with proximal or distal myopathy. Dysferlin is responsible for membrane resealing, and mutations may result in a defect in membrane repair following mechanical or chemical stress, causing an influx of Ca2+. Since mitochondria are involved in Ca2+ buffering, we hypothesised that mitochondrial defects may be present in skeletal muscle biopsies from patients with mutations in this gene. The aim was to characterise mitochondrial defects in muscle from patients with dysferlinopathies. Here, we analysed skeletal muscle biopsies for eight patients by quadruple immunofluorescent assay to assess oxidative phosphorylation protein abundance. Long-range PCR in single muscle fibres was used to look for presence of clonally expanded large-scale mitochondrial DNA rearrangements in patients' skeletal muscle (n = 3). Immunofluorescence demonstrated that the percentage of complex I- and complex IV-deficient fibres was higher in patients with DYSF mutations than in age-matched controls. No clonally expanded mtDNA deletions were detected using long-range PCR in any of the analysed muscle fibres. We conclude that complex I and complex IV deficiency is higher in patients than age matched controls but patients do not have rearrangements of the mtDNA. We hypothesise that respiratory chain deficiency may be the results of an increased cytosolic Ca2+ concentration (due to a membrane resealing defect) causing mitochondrial aberrations.
Collapse
Affiliation(s)
- Amy E Vincent
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Hannah S Rosa
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Charlotte L Alston
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - John P Grady
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Karolina A Rygiel
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Mariana C Rocha
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Rita Barresi
- Rare Diseases Advisory Group Service for Rare Neuromuscular Diseases, Muscle Immunoanalysis Unit, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE2 4AZ, UK
| | - Robert W Taylor
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Doug M Turnbull
- Wellcome Trust Centre for Mitochondrial Research, Institute of Neuroscience, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
43
|
Konkay K, Kannan MA, Lingappa L, Uppin MS, Challa S. Congenital muscular dystrophy with inflammation: Diagnostic considerations. Ann Indian Acad Neurol 2016; 19:356-9. [PMID: 27570388 PMCID: PMC4980959 DOI: 10.4103/0972-2327.186814] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Background and Purpose: Muscle biopsy features of congenital muscular dystrophies (CMD) vary from usual dystrophic picture to normal or nonspecific myopathic picture or prominent fibrosis or striking inflammatory infiltrate, which may lead to diagnostic errors. A series of patients of CMD with significant inflammatory infiltrates on muscle biopsy were correlated with laminin α2 deficiency on immunohistochemistry (IHC). Material and Methods: Cryostat sections of muscle biopsies from the patients diagnosed as CMD on clinical and muscle biopsy features from 1996 to 2014 were reviewed with hematoxylin and eosin(H&E), enzyme and immunohistochemistry (IHC) with laminin α2. Muscle biopsies with inflammatory infiltrate were correlated with laminin α2 deficiency. Results: There were 65 patients of CMD, with inflammation on muscle biopsy in 16. IHC with laminin α2 was available in nine patients, of which six showed complete absence along sarcolemma (five presented with floppy infant syndrome and one with delayed motor milestones) and three showed discontinuous, and less intense staining. Conclusions: CMD show variable degrees of inflammation on muscle biopsy. A diagnosis of laminin α2 deficient CMD should be considered in patients of muscular dystrophy with inflammation, in children with hypotonia/delayed motor milestones.
Collapse
Affiliation(s)
- Kaumudi Konkay
- Department of Pathology, Guntur Medical College, Guntur, Andhra Pradesh, India
| | - Meena Angamuthu Kannan
- Department of Neurology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Lokesh Lingappa
- Department of Paediatric Neurology, Rainbow Hospitals, Hyderabad, Telangana, India
| | - Megha S Uppin
- Department of Pathology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| | - Sundaram Challa
- Department of Pathology, Nizam's Institute of Medical Sciences, Hyderabad, Telangana, India
| |
Collapse
|
44
|
Durward-Akhurst SA, Finno CJ, Barnes N, Shivers J, Guo LT, Shelton GD, Valberg SJ. Major Histocompatibility Complex I and II Expression and Lymphocytic Subtypes in Muscle of Horses with Immune-Mediated Myositis. J Vet Intern Med 2016; 30:1313-21. [PMID: 27352021 PMCID: PMC5094553 DOI: 10.1111/jvim.14371] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Revised: 03/25/2016] [Accepted: 06/11/2016] [Indexed: 11/28/2022] Open
Abstract
BACKGROUND Major histocompatibility complex (MHC) I and II expression is not normally detected on sarcolemma, but is detected with lymphocytic infiltrates in immune-mediated myositis (IMM) of humans and dogs and in dysferlin-deficient muscular dystrophy. HYPOTHESIS/OBJECTIVES To determine if sarcolemmal MHC is expressed in active IMM in horses, if MHC expression is associated with lymphocytic subtype, and if dysferlin is expressed in IMM. ANIMALS Twenty-one IMM horses of Quarter Horse-related breeds, 3 healthy and 6 disease controls (3 pasture myopathy, 3 amylase-resistant polysaccharide storage myopathy [PSSM]). METHODS Immunohistochemical staining for MHC I, II, and CD4+, CD8+, CD20+ lymphocytes was performed on archived muscle of IMM and control horses. Scores were given for MHC I, II, and lymphocytic subtypes. Immunofluorescent staining for dysferlin, dystrophin, and a-sarcoglycan was performed. RESULTS Sarcolemmal MHC I and II expression was detected in 17/21 and 15/21 of IMM horses, respectively, and in specific fibers of PSSM horses, but not healthy or pasture myopathy controls. The CD4+, CD8+, and CD20+ cells were present in 20/21 IMM muscles with CD4+ predominance in 10/21 and CD8+ predominance in 6/21 of IMM horses. Dysferlin, dystrophin, and a-sarcoglycan staining were similar in IMM and control muscles. CONCLUSIONS AND CLINICAL IMPORTANCE Deficiencies of dysferlin, dystrophin, and a-sarcoglycan are not associated with IMM. Sarcolemmal MHC I and II expression in a proportion of myofibers of IMM horses in conjunction with lymphocytic infiltration supports an immune-mediated etiology for IMM. The MHC expression also occured in specific myofibers in PSSM horses in the absence of lymphocytic infiltrates.
Collapse
Affiliation(s)
- S A Durward-Akhurst
- Department of Veterinary Population Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN
| | - C J Finno
- Department of Population Health and Reproduction, University of California-Davis, Davis, CA
| | - N Barnes
- Department of Veterinary Population Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN
| | - J Shivers
- Department of Veterinary Population Sciences, College of Veterinary Medicine, University of Minnesota, St. Paul, MN
| | - L T Guo
- Department of Pathology, University of California, San Diego, La Jolla, CA
| | - G D Shelton
- Department of Pathology, University of California, San Diego, La Jolla, CA
| | - S J Valberg
- Department of Large Animal Clinical Sciences, Michigan State University, East Lansing, MI
| |
Collapse
|
45
|
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: 26] [Impact Index Per Article: 2.9] [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
|
46
|
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
|
47
|
[Dysferlinopathy masquerading as a refractory polymyositis]. Med Clin (Barc) 2015; 145:414-5. [PMID: 25662721 DOI: 10.1016/j.medcli.2014.12.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 11/23/2022]
|
48
|
Cohen TV, Many GM, Fleming BD, Gnocchi VF, Ghimbovschi S, Mosser DM, Hoffman EP, Partridge TA. Upregulated IL-1β in dysferlin-deficient muscle attenuates regeneration by blunting the response to pro-inflammatory macrophages. Skelet Muscle 2015; 5:24. [PMID: 26251696 PMCID: PMC4527226 DOI: 10.1186/s13395-015-0048-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 06/16/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Loss-of-function mutations in the dysferlin gene (DYSF) result in a family of muscle disorders known collectively as the dysferlinopathies. Dysferlin-deficient muscle is characterized by inflammatory foci and macrophage infiltration with subsequent decline in muscle function. Whereas macrophages function to remove necrotic tissue in acute injury, their prevalence in chronic myopathy is thought to inhibit resolution of muscle regeneration. Two major classes of macrophages, classical (M1) and alternative (M2a), play distinct roles during the acute injury process. However, their individual roles in chronic myopathy remain unclear and were explored in this study. METHODS To test the roles of the two macrophage phenotypes on regeneration in dysferlin-deficient muscle, we developed an in vitro co-culture model of macrophages and muscle cells. We assayed the co-cultures using ELISA and cytokine arrays to identify secreted factors and performed transcriptome analysis of molecular networks induced in the myoblasts. RESULTS Dysferlin-deficient muscle contained an excess of M1 macrophage markers, compared with WT, and regenerated poorly in response to toxin injury. Co-culturing macrophages with muscle cells showed that M1 macrophages inhibit muscle regeneration whereas M2a macrophages promote it, especially in dysferlin-deficient muscle cells. Examination of soluble factors released in the co-cultures and transcriptome analysis implicated two soluble factors in mediating the effects: IL-1β and IL-4, which during acute injury are secreted from M1 and M2a macrophages, respectively. To test the roles of these two factors in dysferlin-deficient muscle, myoblasts were treated with IL-4, which improved muscle differentiation, or IL-1β, which inhibited it. Importantly, blockade of IL-1β signaling significantly improved differentiation of dysferlin-deficient cells. CONCLUSIONS We propose that the inhibitory effects of M1 macrophages on myogenesis are mediated by IL-1β signals and suppression of the M1-mediated immune response may improve muscle regeneration in dysferlin deficiency. Our studies identify a potential therapeutic approach to promote muscle regeneration in dystrophic muscle.
Collapse
Affiliation(s)
- Tatiana V. Cohen
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
- />Center for Genetic Muscle Disorders, Kennedy Krieger Institute, 707 N. Broadway, Baltimore, MD 21205 USA
- />Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Gina M. Many
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - Bryan D. Fleming
- />Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742 USA
| | - Viola F. Gnocchi
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - Svetlana Ghimbovschi
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - David M. Mosser
- />Department of Cell Biology and Molecular Genetics, University of Maryland, College Park, MD 20742 USA
| | - Eric P. Hoffman
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| | - Terence A. Partridge
- />Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue NW, Washington, DC 20010 USA
| |
Collapse
|
49
|
Findlay AR, Goyal NA, Mozaffar T. An overview of polymyositis and dermatomyositis. Muscle Nerve 2015; 51:638-56. [PMID: 25641317 DOI: 10.1002/mus.24566] [Citation(s) in RCA: 151] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2015] [Indexed: 12/23/2022]
Abstract
Polymyositis and dermatomyositis are inflammatory myopathies that differ in their clinical features, histopathology, response to treatment, and prognosis. Although their clinical pictures differ, they both present with symmetrical, proximal muscle weakness. Treatment relies mainly upon empirical use of corticosteroids and immunosuppressive agents. A deeper understanding of the molecular pathways that drive pathogenesis, careful phenotyping, and accurate disease classification will aid clinical research and development of more efficacious treatments. In this review we address the current knowledge of the epidemiology, clinical characteristics, diagnostic evaluation, classification, pathogenesis, treatment, and prognosis of polymyositis and dermatomyositis.
Collapse
Affiliation(s)
- Andrew R Findlay
- Department of Neurology, University of California, Irvine UC Irvine, MDA ALS and Neuromuscular Center, 200 South Manchester Avenue, Suite 110, Orange, California, 92868, USA
| | | | | |
Collapse
|
50
|
Dillingham BC, Benny Klimek ME, Gernapudi R, Rayavarapu S, Gallardo E, Van der Meulen JH, Jordan S, Ampong B, Gordish-Dressman H, Spurney CF, Nagaraju K. Inhibition of inflammation with celastrol fails to improve muscle function in dysferlin-deficient A/J mice. J Neurol Sci 2015; 356:157-62. [PMID: 26119397 DOI: 10.1016/j.jns.2015.06.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Revised: 06/18/2015] [Accepted: 06/22/2015] [Indexed: 01/17/2023]
Abstract
The dysferlin-deficient A/J mouse strain represents a homologous model for limb-girdle muscular dystrophy 2B. We evaluated the disease phenotype in 10 month old A/J mice compared to two dysferlin-sufficient, C57BL/6 and A/JOlaHsd, mouse lines to determine which functional end-points are sufficiently sensitive to define the disease phenotype for use in preclinical studies in the A/J strain. A/J mice had significantly lower open field behavioral activity (horizontal activity, total distance, movement time and vertical activity) when compared to C57BL/6 and A/JoIaHsd mice. Both A/J and A/JOIaHsd mice showed decreases in latency to fall with rotarod compared to C57BL/6. No changes were detected in grip strength, force measurements or motor coordination between these three groups. Furthermore, we have found that A/J muscle shows significantly increased levels of the pro-inflammatory cytokine TNF-α when compared to C57BL/6 mice, indicating an activation of NF-κB signaling as part of the inflammatory response in dysferlin-deficient muscle. Therefore, we assessed the effect of celastrol (a potent NF-κB inhibitor) on the disease phenotype in female A/J mice. Celastrol treatment for four months significantly reduced the inflammation in A/J muscle; however, it had no beneficial effect in improving muscle function, as assessed by grip strength, open field activity, and in vitro force contraction. In fact, celastrol treated mice showed a decrease in body mass, hindlimb grip strength and maximal EDL force. These findings suggest that inhibition of inflammation alone may not be sufficient to improve the muscle disease phenotype in dysferlin-deficient mice and may require combination therapies that target membrane stability to achieve a functional improvement in skeletal muscle.
Collapse
Affiliation(s)
- Blythe C Dillingham
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington, D.C., USA
| | - Margaret E Benny Klimek
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington, D.C., USA
| | - Ramkishore Gernapudi
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington, D.C., USA
| | - Sree Rayavarapu
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington, D.C., USA
| | - Eduard Gallardo
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Jack H Van der Meulen
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Sarah Jordan
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Beryl Ampong
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Heather Gordish-Dressman
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Christopher F Spurney
- Institut de Recerca Hospital de la Santa Creu i Sant Pau. U.A.B.C./Pare Claret, 167 08025 Barcelona, Spain
| | - Kanneboyina Nagaraju
- Research Center for Genetic Medicine, Children's National Medical Center, 111 Michigan Ave NW, Washington, D.C., USA; Department of Integrative Systems Biology, Institute for Biomedical Sciences, The George Washington University, 2300 Eye Street, N.W., Ross 605, Washington, D.C., USA.
| |
Collapse
|