1
|
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
|
2
|
Signorelli M, Ebrahimpoor M, Veth O, Hettne K, Verwey N, García‐Rodríguez R, Tanganyika‐deWinter CL, Lopez Hernandez LB, Escobar Cedillo R, Gómez Díaz B, Magnusson OT, Mei H, Tsonaka R, Aartsma‐Rus A, Spitali P. Peripheral blood transcriptome profiling enables monitoring disease progression in dystrophic mice and patients. EMBO Mol Med 2021; 13:e13328. [PMID: 33751844 PMCID: PMC8033515 DOI: 10.15252/emmm.202013328] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 02/09/2021] [Accepted: 02/10/2021] [Indexed: 12/13/2022] Open
Abstract
DMD is a rare disorder characterized by progressive muscle degeneration and premature death. Therapy development is delayed by difficulties to monitor efficacy non-invasively in clinical trials. In this study, we used RNA-sequencing to describe the pathophysiological changes in skeletal muscle of 3 dystrophic mouse models. We show how dystrophic changes in muscle are reflected in blood by analyzing paired muscle and blood samples. Analysis of repeated blood measurements followed the dystrophic signature at five equally spaced time points over a period of seven months. Treatment with two antisense drugs harboring different levels of dystrophin recovery identified genes associated with safety and efficacy. Evaluation of the blood gene expression in a cohort of DMD patients enabled the comparison between preclinical models and patients, and the identification of genes associated with physical performance, treatment with corticosteroids and body measures. The presented results provide evidence that blood RNA-sequencing can serve as a tool to evaluate disease progression in dystrophic mice and patients, as well as to monitor response to (dystrophin-restoring) therapies in preclinical drug development and in clinical trials.
Collapse
Affiliation(s)
- Mirko Signorelli
- Department of Biomedical Data SciencesLeiden University Medical CenterLeidenThe Netherlands
| | - Mitra Ebrahimpoor
- Department of Biomedical Data SciencesLeiden University Medical CenterLeidenThe Netherlands
| | - Olga Veth
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Kristina Hettne
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | - Nisha Verwey
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| | | | | | - Luz B Lopez Hernandez
- Departamento de Medicina GenómicaUniversidad Autónoma de GuadalajaraGuadalajaraMexico
- Centro Médico Nacional "20 de Noviembre", ISSSTECiudad de MéxicoMexico
| | | | - Benjamín Gómez Díaz
- Sociedad Mexicana de la Distrofia Muscular A.C INR‐LGIICiudad de MéxicoMexico
| | | | - Hailiang Mei
- Sequencing Analysis Support CoreLeiden University Medical CenterLeidenThe Netherlands
| | - Roula Tsonaka
- Department of Biomedical Data SciencesLeiden University Medical CenterLeidenThe Netherlands
| | | | - Pietro Spitali
- Department of Human GeneticsLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
3
|
Muscle Diversity, Heterogeneity, and Gradients: Learning from Sarcoglycanopathies. Int J Mol Sci 2021; 22:ijms22052502. [PMID: 33801487 PMCID: PMC7958856 DOI: 10.3390/ijms22052502] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/25/2021] [Accepted: 02/26/2021] [Indexed: 12/25/2022] Open
Abstract
Skeletal muscle, the most abundant tissue in the body, is heterogeneous. This heterogeneity forms the basis of muscle diversity, which is reflected in the specialized functions of muscles in different parts of the body. However, these different parts are not always clearly delimitated, and this often gives rise to gradients within the same muscle and even across the body. During the last decade, several studies on muscular disorders both in mice and in humans have observed particular distribution patterns of muscle weakness during disease, indicating that the same mutation can affect muscles differently. Moreover, these phenotypical differences reveal gradients of severity, existing alongside other architectural gradients. These two factors are especially prominent in sarcoglycanopathies. Nevertheless, very little is known about the mechanism(s) driving the phenotypic diversity of the muscles affected by these diseases. Here, we will review the available literature on sarcoglycanopathies, focusing on phenotypic differences among affected muscles and gradients, characterization techniques, molecular signatures, and cell population heterogeneity, highlighting the possibilities opened up by new technologies. This review aims to revive research interest in the diverse disease phenotype affecting different muscles, in order to pave the way for new therapeutic interventions.
Collapse
|
4
|
Hiller M, Geissler M, Janssen G, van Veelen P, Aartsma-Rus A, Spitali P. The mRNA Binding Proteome of Proliferating and Differentiated Muscle Cells. GENOMICS PROTEOMICS & BIOINFORMATICS 2020; 18:384-396. [PMID: 33338663 PMCID: PMC8242265 DOI: 10.1016/j.gpb.2020.06.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 02/29/2020] [Accepted: 06/04/2020] [Indexed: 01/24/2023]
Abstract
Muscle formation is a coordinated process driven by extensive gene expression changes where single cells fuse together to form multinucleated muscle fibers. Newly synthesized mRNAs are then regulated by RNA binding proteins (RBPs), affecting post-transcriptional transcript metabolism. Here, we determined how large-scale gene expression changes affect the catalog of RBPs by studying proliferating and differentiated muscle cells in healthy and dystrophic conditions. Transcriptomic analysis showed that the expression of more than 7000 genes was affected during myogenesis. We identified 769 RBPs, of which 294 were muscle-specific and 49 were uniquely shared with cardiomyocytes. A subset of 32 RBPs (half of which were muscle-specific) was found to be preferentially associated with target mRNAs in either myoblasts (MBs) or myotubes (MTs). A large proportion of catalytic proteins were bound to mRNAs even though they lack classical RNA binding domains. Finally, we showed how the identification of cell-specific RBPs enabled the identification of biomarkers that can separate healthy individuals from dystrophic patients. Our data show how interactome data can shed light on new basic RNA biology as well as provide cell-specific data that can be used for diagnostic purposes.
Collapse
Affiliation(s)
- Monika Hiller
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
| | - Miriam Geissler
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
| | - George Janssen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
| | - Peter van Veelen
- Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZC, the Netherlands
| | - Pietro Spitali
- Department of Human Genetics, Leiden University Medical Center, Leiden 2333 ZC, the Netherlands.
| |
Collapse
|
5
|
Verhaart IEC, Putker K, van de Vijver D, Tanganyika-de Winter CL, Pasteuning-Vuhman S, Plomp JJ, Aartsma-Rus AM, van Putten M. Cross-sectional study into age-related pathology of mouse models for limb girdle muscular dystrophy types 2D and 2F. PLoS One 2019; 14:e0220665. [PMID: 31430305 PMCID: PMC6701749 DOI: 10.1371/journal.pone.0220665] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Accepted: 07/19/2019] [Indexed: 12/11/2022] Open
Abstract
Limb girdle muscular dystrophy (LGMD) types 2D and 2F are caused by mutations in the genes encoding for α- and δ-sarcoglycan, respectively, leading to progressive muscle weakness. Mouse models exist for LGMD2D (Sgca-/-) and 2F (Sgcd-/-). In a previous natural history study, we described the pathology in these mice at 34 weeks of age. However, the development of muscle pathology at younger ages has not been fully characterised yet. We therefore performed a study into age-related changes in muscle function and pathology by examining mice at different ages. From 4 weeks of age onwards, male mice were subjected to functional tests and sacrificed at respectively 8, 16 or 24 weeks of age. Muscle histopathology and expression of genes involved in muscle pathology were analysed for several skeletal muscles, while miRNA levels were assessed in serum. In addition, for Sgcd-/- mice heart pathology was assessed. Muscle function showed a gradual decline in both Sgca-/- and Sgcd-/- mice. Respiratory function was also impaired at all examined timepoints. Already at 8 weeks of age, muscle pathology was prominent, and fibrotic, inflammatory and regenerative markers were elevated, which remained relatively constant with age. In addition, Sgcd-/- mice showed signs of cardiomyopathy from 16 weeks of age onwards. These results indicate that Sgca-/- and Sgcd-/- are relevant disease models for LGMD2D and 2F.
Collapse
Affiliation(s)
- Ingrid E. C. Verhaart
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Kayleigh Putker
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Davy van de Vijver
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | | | | | - Jaap J. Plomp
- Department of Neurology, Leiden University Medical Center, Leiden, the Netherlands
| | - Annemieke M. Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
- * E-mail:
| | - Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
6
|
Malatras A, Duguez S, Duddy W. Muscle Gene Sets: a versatile methodological aid to functional genomics in the neuromuscular field. Skelet Muscle 2019; 9:10. [PMID: 31053169 PMCID: PMC6498474 DOI: 10.1186/s13395-019-0196-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Accepted: 04/09/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The approach of building large collections of gene sets and then systematically testing hypotheses across these collections is a powerful tool in functional genomics, both in the pathway analysis of omics data and to uncover the polygenic effects associated with complex diseases in genome-wide association study. The Molecular Signatures Database includes collections of oncogenic and immunologic signatures enabling researchers to compare transcriptional datasets across hundreds of previous studies and leading to important insights in these fields, but such a resource does not currently exist for neuromuscular research. In previous work, we have shown the utility of gene set approaches to understand muscle cell physiology and pathology. METHODS Following a systematic survey of public muscle data, we passed gene expression profiles from 4305 samples through a robust pre-processing and standardized data analysis pipeline. Two hundred eighty-two samples were discarded based on a battery of rigorous global quality controls. From among the remaining studies, 578 comparisons of interest were identified by a combination of text mining and manual curation of the study meta-data. For each comparison, significantly dysregulated genes (FDR adjusted p < 0.05) were identified. RESULTS Lists of dysregulated genes were divided between upregulated and downregulated to give 1156 Muscle Gene Sets (MGS). This resource is available for download ( www.sys-myo.com/muscle_gene_sets ) and is accessible through three commonly used functional genomics platforms (GSEA, EnrichR, and WebGestalt). Basic guidance and recommendations are provided for the use of MGS through these platforms. In addition, consensus muscle gene sets were created to capture the overlap between the results of similar studies, and analysis of these highlighted the potential for novel disease-relevant findings. CONCLUSIONS The MGS resource can be used to investigate the behaviour of any list of genes across previous comparisons of muscle conditions, to compare previous studies to one another, and to explore the functional relationship of muscle dysregulation to the Gene Ontology. Its major intended use is in enrichment testing for functional genomics analysis.
Collapse
Affiliation(s)
- Apostolos Malatras
- Myologie Centre de Recherche, Université Sorbonne, UMRS 974 UPMC, INSERM, FRE 3617 CNRS, AIM, Paris, France
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, C-TRIC, Ulster University, Altnagelvin Hospital Campus, Glenshane Road, Derry/Londonderry, BT47 6SB UK
- Department of Biological Sciences, Molecular Medicine Research Center, University of Cyprus, 1 University Avenue, 2109 Nicosia, Cyprus
| | - Stephanie Duguez
- Myologie Centre de Recherche, Université Sorbonne, UMRS 974 UPMC, INSERM, FRE 3617 CNRS, AIM, Paris, France
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, C-TRIC, Ulster University, Altnagelvin Hospital Campus, Glenshane Road, Derry/Londonderry, BT47 6SB UK
| | - William Duddy
- Myologie Centre de Recherche, Université Sorbonne, UMRS 974 UPMC, INSERM, FRE 3617 CNRS, AIM, Paris, France
- Northern Ireland Centre for Stratified Medicine, Biomedical Sciences Research Institute, C-TRIC, Ulster University, Altnagelvin Hospital Campus, Glenshane Road, Derry/Londonderry, BT47 6SB UK
| |
Collapse
|
7
|
Effects of metformin on congenital muscular dystrophy type 1A disease progression in mice: a gender impact study. Sci Rep 2018; 8:16302. [PMID: 30389963 PMCID: PMC6214987 DOI: 10.1038/s41598-018-34362-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 10/06/2018] [Indexed: 02/08/2023] Open
Abstract
Congenital muscular dystrophy with laminin α2 chain-deficiency (LAMA2-CMD) is a severe muscle disorder with complex underlying pathogenesis. We have previously employed profiling techniques to elucidate molecular patterns and demonstrated significant metabolic impairment in skeletal muscle from LAMA2-CMD patients and mouse models. Thus, we hypothesize that skeletal muscle metabolism may be a promising pharmacological target to improve muscle function in LAMA2-CMD. Here, we have investigated whether the multifunctional medication metformin could be used to reduce disease in the dy2J/dy2J mouse model of LAMA2-CMD. First, we show gender disparity for several pathological hallmarks of LAMA2-CMD. Second, we demonstrate that metformin treatment significantly increases weight gain and energy efficiency, enhances muscle function and improves skeletal muscle histology in female dy2J/dy2J mice (and to a lesser extent in dy2J/dy2J males). Thus, our current data suggest that metformin may be a potential future supportive treatment that improves many of the pathological characteristics of LAMA2-CMD.
Collapse
|
8
|
Gawor M, Prószyński TJ. The molecular cross talk of the dystrophin-glycoprotein complex. Ann N Y Acad Sci 2017; 1412:62-72. [DOI: 10.1111/nyas.13500] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 08/29/2017] [Accepted: 09/04/2017] [Indexed: 12/25/2022]
Affiliation(s)
- Marta Gawor
- Laboratory of Synaptogenesis; Nencki Institute of Experimental Biology; Polish Academy of Sciences Warsaw Poland
| | - Tomasz J. Prószyński
- Laboratory of Synaptogenesis; Nencki Institute of Experimental Biology; Polish Academy of Sciences Warsaw Poland
| |
Collapse
|
9
|
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
|
10
|
Quattrocelli M, Capote J, Ohiri JC, Warner JL, Vo AH, Earley JU, Hadhazy M, Demonbreun AR, Spencer MJ, McNally EM. Genetic modifiers of muscular dystrophy act on sarcolemmal resealing and recovery from injury. PLoS Genet 2017; 13:e1007070. [PMID: 29065150 PMCID: PMC5669489 DOI: 10.1371/journal.pgen.1007070] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Revised: 11/03/2017] [Accepted: 10/11/2017] [Indexed: 12/17/2022] Open
Abstract
Genetic disruption of the dystrophin complex produces muscular dystrophy characterized by a fragile muscle plasma membrane leading to excessive muscle degeneration. Two genetic modifiers of Duchenne Muscular Dystrophy implicate the transforming growth factor β (TGFβ) pathway, osteopontin encoded by the SPP1 gene and latent TGFβ binding protein 4 (LTBP4). We now evaluated the functional effect of these modifiers in the context of muscle injury and repair to elucidate their mechanisms of action. We found that excess osteopontin exacerbated sarcolemmal injury, and correspondingly, that loss of osteopontin reduced injury extent both in isolated myofibers and in muscle in vivo. We found that ablation of osteopontin was associated with reduced expression of TGFβ and TGFβ-associated pathways. We identified that increased TGFβ resulted in reduced expression of Anxa1 and Anxa6, genes encoding key components of the muscle sarcolemma resealing process. Genetic manipulation of Ltbp4 in dystrophic muscle also directly modulated sarcolemmal resealing, and Ltbp4 alleles acted in concert with Anxa6, a distinct modifier of muscular dystrophy. These data provide a model in which a feed forward loop of TGFβ and osteopontin directly impacts the capacity of muscle to recover from injury, and identifies an intersection of genetic modifiers on muscular dystrophy.
Collapse
MESH Headings
- Animals
- Annexin A1/genetics
- Annexin A1/metabolism
- Annexin A6/genetics
- Annexin A6/metabolism
- Female
- Gene Expression Regulation
- Genes, Modifier
- Latent TGF-beta Binding Proteins/physiology
- Male
- Mice
- Mice, Inbred DBA
- Mice, Knockout
- Muscle, Skeletal/injuries
- Muscle, Skeletal/physiology
- Muscular Dystrophy, Animal/genetics
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Animal/pathology
- Osteopontin/genetics
- Osteopontin/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Recovery of Function
- Sarcolemma/physiology
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Joanna Capote
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Joyce C. Ohiri
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - James L. Warner
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Andy H. Vo
- Committee on Development, Regeneration, and Stem Cell Biology, The University of Chicago, Chicago, Illinois, United States of America
| | - Judy U. Earley
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Alexis R. Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| | - Melissa J. Spencer
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, California, United States of America
| | - Elizabeth M. McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States of America
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW To discuss current knowledge on the role of connexins and pannexins in the musculoskeletal system. RECENT FINDINGS Connexins and pannexins are crucial for the development and maintenance of both bone and skeletal muscle. In bone, the presence of connexin and more recently of pannexin channels in osteoblasts, osteoclasts, and osteocytes has been described and shown to be essential for normal skeletal development and bone adaptation. In skeletal muscles, connexins and pannexins play important roles during development and regeneration through coordinated regulation of metabolic functions via cell-to-cell communication. Further, under pathological conditions, altered expression of these proteins can promote muscle atrophy and degeneration by stimulating inflammasome activity. In this review, we highlight the important roles of connexins and pannexins in the development, maintenance, and regeneration of musculoskeletal tissues, with emphasis on the mechanisms by which these molecules mediate chemical (e.g., ATP and prostaglandin E2) and physical (e.g., mechanical stimulation) stimuli that target the musculoskeletal system and their involvement in the pathophysiological changes in both genetic and acquired diseases.
Collapse
Affiliation(s)
- Lilian I Plotkin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS5045, Indianapolis, IN, 46202, USA.
- Roudebush Veterans Administration Medical Center, Indianapolis, Indiana, USA.
- Indiana Center for Musculoskeletal Health, Indianapolis, Indiana, USA.
| | - Hannah M Davis
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, 635 Barnhill Drive, MS5045, Indianapolis, IN, 46202, USA
| | - Bruno A Cisterna
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Alameda 340, Santiago, Chile
| | - Juan C Sáez
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Alameda 340, Santiago, Chile.
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
12
|
Quattrocelli M, Spencer MJ, McNally EM. Outside in: The matrix as a modifier of muscular dystrophy. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2017; 1864:572-579. [PMID: 28011285 PMCID: PMC5262521 DOI: 10.1016/j.bbamcr.2016.12.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Revised: 12/14/2016] [Accepted: 12/19/2016] [Indexed: 02/07/2023]
Abstract
Muscular dystrophies are genetic conditions leading to muscle degeneration and often, impaired regeneration. Duchenne Muscular Dystrophy is a prototypical form of muscular dystrophy, and like other forms of genetically inherited muscle diseases, pathological progression is variable. Variability in muscular dystrophy can arise from differences in the manner in which the primary mutation impacts the affected protein's function; however, clinical heterogeneity also derives from secondary mutations in other genes that can enhance or reduce pathogenic features of disease. These genes, called genetic modifiers, regulate the pathophysiological context of dystrophic degeneration and regeneration. Understanding the mechanistic links between genetic modifiers and dystrophic progression sheds light on pathologic remodeling, and provides novel avenues to therapeutically intervene to reduce muscle degeneration. Based on targeted genetic approaches and unbiased genomewide screens, several modifiers have been identified for muscular dystrophy, including extracellular agonists of signaling cascades. This review will focus on identification and possible mechanisms of recently identified modifiers for muscular dystrophy, including osteopontin, latent TGFβ binding protein 4 (LTBP4) and Jagged1. Moreover, we will review the investigational approaches that aim to target modifier pathways and thereby counteract dystrophic muscle wasting.
Collapse
Affiliation(s)
| | - Melissa J Spencer
- Dept of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | |
Collapse
|
13
|
Hogarth MW, Houweling PJ, Thomas KC, Gordish-Dressman H, Bello L, Pegoraro E, Hoffman EP, Head SI, North KN. Evidence for ACTN3 as a genetic modifier of Duchenne muscular dystrophy. Nat Commun 2017; 8:14143. [PMID: 28139640 PMCID: PMC5290331 DOI: 10.1038/ncomms14143] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Accepted: 11/22/2016] [Indexed: 01/01/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by muscle degeneration and progressive weakness. There is considerable inter-patient variability in disease onset and progression, which can confound the results of clinical trials. Here we show that a common null polymorphism (R577X) in ACTN3 results in significantly reduced muscle strength and a longer 10 m walk test time in young, ambulant patients with DMD; both of which are primary outcome measures in clinical trials. We have developed a double knockout mouse model, which also shows reduced muscle strength, but is protected from stretch-induced eccentric damage with age. This suggests that α-actinin-3 deficiency reduces muscle performance at baseline, but ameliorates the progression of dystrophic pathology. Mechanistically, we show that α-actinin-3 deficiency triggers an increase in oxidative muscle metabolism through activation of calcineurin, which likely confers the protective effect. Our studies suggest that ACTN3 R577X genotype is a modifier of clinical phenotype in DMD patients.
Collapse
Affiliation(s)
- Marshall W Hogarth
- Institute for Neuroscience and Muscle Research, The Children's Hospital Westmead, New South Wales 2145, Australia.,Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, New South Wales 2006, Australia
| | - Peter J Houweling
- Institute for Neuroscience and Muscle Research, The Children's Hospital Westmead, New South Wales 2145, Australia.,School of Medical Sciences, University of New South Wales, New South Wales 2052, Australia.,Murdoch Childrens Research Institute, Melbourne, Victoria 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria 3010, Australia
| | - Kristen C Thomas
- Institute for Neuroscience and Muscle Research, The Children's Hospital Westmead, New South Wales 2145, Australia
| | - Heather Gordish-Dressman
- Research Centre for Genetic Medicine, Children's National Medical Centre, Washington DC 20010, USA
| | - Luca Bello
- Research Centre for Genetic Medicine, Children's National Medical Centre, Washington DC 20010, USA.,Department of Neurosciences, University of Padova, Padova 35122, Italy
| | | | - Elena Pegoraro
- Department of Neurosciences, University of Padova, Padova 35122, Italy
| | - Eric P Hoffman
- Research Centre for Genetic Medicine, Children's National Medical Centre, Washington DC 20010, USA
| | - Stewart I Head
- School of Medical Sciences, University of New South Wales, New South Wales 2052, Australia
| | - Kathryn N North
- Institute for Neuroscience and Muscle Research, The Children's Hospital Westmead, New South Wales 2145, Australia.,Discipline of Paediatrics and Child Health, Faculty of Medicine, University of Sydney, New South Wales 2006, Australia.,Murdoch Childrens Research Institute, Melbourne, Victoria 3052, Australia.,Department of Paediatrics, Faculty of Medicine, Dentistry and Health Sciences, University of Melbourne, Melbourne, Victoria 3010, Australia
| |
Collapse
|
14
|
Abstract
PURPOSE OF REVIEW Recently, genetic pathways that modify the clinical severity of Duchenne muscular dystrophy (DMD) have been identified. The pathways uncovered as modifiers are useful to predict prognosis and also elucidate molecular signatures that can be manipulated therapeutically. RECENT FINDINGS Modifiers have been identified using combinations of transcriptome and genome profiling. Osteopontin, encoded by the SPP1 gene, was found using gene expression profiling. Latent TGFβ binding protein 4, encoding latent TGFβ binding protein 4 was initially discovered using a genome-wide screen in mice and then validated in cohorts of DMD patients. These two pathways converge in that they both regulate TGFβ. A third modifier, Anxa6 that specifies annexin A6, is a calcium binding protein that has been identified using mouse models, and regulates the injury pathway and sarcolemmal resealing. SUMMARY Genetic modifiers can serve as biomarkers for outcomes in DMD. Modifiers can alter strength and ambulation in muscular dystrophy, and these same features can be used as endpoints used in clinical trials. Moreover, because genetic modifiers can influence outcomes, these genetic markers should be considered when stratifying results in muscular dystrophy.
Collapse
Affiliation(s)
- Andy H Vo
- Committee on Development, Regeneration and Stem Cell Biology, Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, The University of Chicago, Chicago, Illinois, USA
| | | |
Collapse
|
15
|
Ferreira B, Da Silva GP, Gonçalves CR, Arnoni VW, Siéssere S, Semprini M, Verri ED, Chaves TC, Regalo SCH. Stomatognathic function in Duchenne muscular dystrophy: a case-control study. Dev Med Child Neurol 2016; 58:516-21. [PMID: 26991937 DOI: 10.1111/dmcn.13094] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/14/2016] [Indexed: 11/27/2022]
Abstract
AIM This study aimed to analyse electromyographic activity, masticatory efficiency, muscle thickness, and bite force of individuals with Duchenne muscular dystrophy (DMD). METHOD Forty males aged 4-15 years, 20 with DMD and 20 healthy age-, height-, and weight-matched controls, underwent electromyography and ultrasonography of temporalis, masseter, and sternocleidomastoid muscles during postural control of the jaw, mastication, and maximal molar bite force. RESULTS The normalized electromyography signals showed higher activity in masseter and temporal muscles at rest, during protrusion, left and right laterality, and fatigue condition in the group with DMD than in the comparison group (p≤0.05). For masticatory efficiency of cycles, in analysis of non-habitual chewing of flavourless gum, and habitual chewing of peanuts and raisins, the group with DMD presented lower averages (p≤0.05). For the muscle thickness, the results showed that there was a lower muscle thickness in the group with DMD for all muscles during the rest and maximal voluntary contraction, except for masseter and sternocleidomastoid in the maximal voluntary contraction. In the maximal molar bite force, the group with DMD presented higher values for both sides than the comparison group (p≤0.05). INTERPRETATION Patients with DMD show muscle changes related to the stomatognathic system, in their activity, bite force, and muscle thickness.
Collapse
Affiliation(s)
- Bruno Ferreira
- Department of Morphology, Physiology and Basic Pathology, University of São Paulo, Ribeirão Preto, Brazil
| | - Gabriel Pádua Da Silva
- Department of Morphology, Physiology and Basic Pathology, University of São Paulo, Ribeirão Preto, Brazil
| | - Camila Rosa Gonçalves
- Department of Morphology, Physiology and Basic Pathology, University of São Paulo, Ribeirão Preto, Brazil
| | - Veridiana Wanshi Arnoni
- Department of Morphology, Physiology and Basic Pathology, University of São Paulo, Ribeirão Preto, Brazil
| | - Selma Siéssere
- Department of Morphology, Physiology and Basic Pathology, University of São Paulo, Ribeirão Preto, Brazil
| | - Marisa Semprini
- Department of Morphology, Physiology and Basic Pathology, University of São Paulo, Ribeirão Preto, Brazil
| | - Edson Donizetti Verri
- Department of Morphology, Physiology and Basic Pathology, University of São Paulo, Ribeirão Preto, Brazil
| | - Thais Cristina Chaves
- Department of Neuroscience and Behavior, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | | |
Collapse
|
16
|
Retracted: 'The effects of Duchenne muscular dystrophy on the performance of the stomatognathic system: case-control study', by Ferreira B., Da Silva G.P., Gonçalves C.R., et al. Dev Med Child Neurol 2016; 58:523. [PMID: 25683793 DOI: 10.1111/dmcn.12707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The above article, published online on 13 February 2015 in Wiley Online Library Early View (wileyonlinelibrary.com), has been retracted by agreement between the authors, the journal editors, Bernard Dan and Peter Rosenbaum, the MacKeith Press and John Wiley & Sons Ltd. The retraction has been made due to concerns relating to the validity of the EMG methodology and its interpretation, which affect the results of the paper. Reference Ferreira B, Da Silva GP, Gonçalves CR, Arnoni VW, Siéssere S, Semprini M, Verri ED, Chaves TC, Regalo SH. The effects of Duchenne muscular dystrophy on the performance of the stomatognathic system: case-control study. Developmental Medicine & Child Neurology 2015. doi:10.1111/dmcn.12707.
Collapse
|
17
|
Turk R, Hsiao JJ, Smits MM, Ng BH, Pospisil TC, Jones KS, Campbell KP, Wright ME. Molecular Signatures of Membrane Protein Complexes Underlying Muscular Dystrophy. Mol Cell Proteomics 2016; 15:2169-85. [PMID: 27099343 PMCID: PMC5083101 DOI: 10.1074/mcp.m116.059188] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Indexed: 01/16/2023] Open
Abstract
Mutations in genes encoding components of the sarcolemmal dystrophin-glycoprotein complex (DGC) are responsible for a large number of muscular dystrophies. As such, molecular dissection of the DGC is expected to both reveal pathological mechanisms, and provides a biological framework for validating new DGC components. Establishment of the molecular composition of plasma-membrane protein complexes has been hampered by a lack of suitable biochemical approaches. Here we present an analytical workflow based upon the principles of protein correlation profiling that has enabled us to model the molecular composition of the DGC in mouse skeletal muscle. We also report our analysis of protein complexes in mice harboring mutations in DGC components. Bioinformatic analyses suggested that cell-adhesion pathways were under the transcriptional control of NFκB in DGC mutant mice, which is a finding that is supported by previous studies that showed NFκB-regulated pathways underlie the pathophysiology of DGC-related muscular dystrophies. Moreover, the bioinformatic analyses suggested that inflammatory and compensatory mechanisms were activated in skeletal muscle of DGC mutant mice. Additionally, this proteomic study provides a molecular framework to refine our understanding of the DGC, identification of protein biomarkers of neuromuscular disease, and pharmacological interrogation of the DGC in adult skeletal muscle https://www.mda.org/disease/congenital-muscular-dystrophy/research.
Collapse
Affiliation(s)
- Rolf Turk
- From the ‡Howard Hughes Medical Institute, §Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, ¶Department of Molecular Physiology and Biophysics, ‖Department of Neurology, **Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | | | | | - Brandon H Ng
- ¶Department of Molecular Physiology and Biophysics
| | - Tyler C Pospisil
- From the ‡Howard Hughes Medical Institute, §Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, ¶Department of Molecular Physiology and Biophysics, ‖Department of Neurology, **Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Kayla S Jones
- From the ‡Howard Hughes Medical Institute, §Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, ¶Department of Molecular Physiology and Biophysics, ‖Department of Neurology, **Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Kevin P Campbell
- From the ‡Howard Hughes Medical Institute, §Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, ¶Department of Molecular Physiology and Biophysics, ‖Department of Neurology, **Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | | |
Collapse
|
18
|
Almeida CF, Martins PC, Vainzof M. Comparative transcriptome analysis of muscular dystrophy models Large(myd), Dmd(mdx)/Large(myd) and Dmd(mdx): what makes them different? Eur J Hum Genet 2016; 24:1301-9. [PMID: 26932192 DOI: 10.1038/ejhg.2016.16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Revised: 01/26/2016] [Accepted: 02/01/2016] [Indexed: 11/09/2022] Open
Abstract
Muscular dystrophies (MD) are a clinically and genetically heterogeneous group of Mendelian diseases. The underlying pathophysiology and phenotypic variability in each form are much more complex, suggesting the involvement of many other genes. Thus, here we studied the whole genome expression profile in muscles from three mice models for MD, at different time points: Dmd(mdx) (mutation in dystrophin gene), Large(myd-/-) (mutation in Large) and Dmd(mdx)/Large(myd-/-) (both mutations). The identification of altered biological functions can contribute to understand diseases and to find prognostic biomarkers and points for therapeutic intervention. We identified a substantial number of differentially expressed genes (DEGs) in each model, reflecting diseases' complexity. The main biological process affected in the three strains was immune system, accounting for the majority of enriched functional categories, followed by degeneration/regeneration and extracellular matrix remodeling processes. The most notable differences were in 21-day-old Dmd(mdx), with a high proportion of DEGs related to its regenerative capacity. A higher number of positive embryonic myosin heavy chain (eMyHC) fibers confirmed this. The new Dmd(mdx)/Large(myd-/-) model did not show a highly different transcriptome from the parental lineages, with a profile closer to Large(myd-/-), but not bearing the same regenerative potential as Dmd(mdx). This is the first report about transcriptome profile of a mouse model for congenital MD and Dmd(mdx)/Large(myd). By comparing the studied profiles, we conclude that alterations in biological functions due to the dystrophic process are very similar, and that the intense regeneration in Dmd(mdx) involves a large number of activated genes, not differentially expressed in the other two strains.
Collapse
Affiliation(s)
- Camila F Almeida
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Poliana Cm Martins
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| | - Mariz Vainzof
- Laboratory of Muscle Proteins and Comparative Histopathology, Human Genome and Stem Cell Research Center, Department of Genetics and Evolutionary Biology, Biosciences Institute, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Pelosi L, Berardinelli MG, Forcina L, Spelta E, Rizzuto E, Nicoletti C, Camilli C, Testa E, Catizone A, De Benedetti F, Musarò A. Increased levels of interleukin-6 exacerbate the dystrophic phenotype in mdx mice. Hum Mol Genet 2015; 24:6041-53. [PMID: 26251044 PMCID: PMC4599671 DOI: 10.1093/hmg/ddv323] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 08/03/2015] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is characterized by progressive lethal muscle degeneration and chronic inflammatory response. The mdx mouse strain has served as the animal model for human DMD. However, while DMD patients undergo extensive necrosis, the affected muscles of adult mdx mice rapidly regenerates and regains structural and functional integrity. The basis for the mild effects observed in mice compared with the lethal consequences in humans remains unknown. In this study, we provide evidence that interleukin-6 (IL-6) is causally linked to the pathogenesis of muscular dystrophy. We report that forced expression of IL-6, in the adult mdx mice, recapitulates the severe phenotypic characteristics of DMD in humans. Increased levels of IL-6 exacerbate the dystrophic muscle phenotype, sustaining inflammatory response and repeated cycles of muscle degeneration and regeneration, leading to exhaustion of satellite cells. The mdx/IL6 mouse closely approximates the human disease and more faithfully recapitulates the disease progression in humans. This study promises to significantly advance our understanding of the pathogenic mechanisms that lead to DMD.
Collapse
Affiliation(s)
- Laura Pelosi
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | | | - Laura Forcina
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | - Elisa Spelta
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | - Emanuele Rizzuto
- Department of Mechanical and Aerospace Engineering, Sapienza University of Rome, Rome 00184, Italy
| | - Carmine Nicoletti
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | - Carlotta Camilli
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | - Erika Testa
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and
| | - Angela Catizone
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Rome 00161, Italy
| | | | - Antonio Musarò
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM and Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome 00161, Italy
| |
Collapse
|
20
|
Wiktorowicz T, Kinter J, Kobuke K, Campbell KP, Sinnreich M. Genetic characterization and improved genotyping of the dysferlin-deficient mouse strain Dysf (tm1Kcam). Skelet Muscle 2015; 5:32. [PMID: 26464793 PMCID: PMC4603641 DOI: 10.1186/s13395-015-0057-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2015] [Accepted: 09/03/2015] [Indexed: 11/10/2022] Open
Abstract
Background Mouse models of dysferlinopathies are valuable tools with which to investigate the pathomechanisms underlying these diseases and to test novel therapeutic strategies. One such mouse model is the Dysftm1Kcam strain, which was generated using a targeting vector to replace a 12-kb region of the dysferlin gene and which features a progressive muscular dystrophy. A prerequisite for successful animal studies using genetic mouse models is an accurate genotyping protocol. Unfortunately, the lack of robustness of currently available genotyping protocols for the Dysftm1Kcam mouse has prevented efficient colony management. Initial attempts to improve the genotyping protocol based on the published genomic structure failed. These difficulties led us to analyze the targeted locus of the dysferlin gene of the Dysftm1Kcam mouse in greater detail. Methods In this study we resequenced and analyzed the targeted locus of the Dysftm1Kcam mouse and developed a novel PCR protocol for genotyping. Results We found that instead of a deletion, the dysferlin locus in the Dysftm1Kcam mouse carries a targeted insertion. This genetic characterization enabled us to establish a reliable method for genotyping of the Dysftm1Kcam mouse, and thus has made efficient colony management possible. Conclusion Our work will make the Dysftm1Kcam mouse model more attractive for animal studies of dysferlinopathies. Electronic supplementary material The online version of this article (doi:10.1186/s13395-015-0057-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Tatiana Wiktorowicz
- Neuromuscular Research Group, Departments of Neurology and Biomedicine, University and University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Jochen Kinter
- Neuromuscular Research Group, Departments of Neurology and Biomedicine, University and University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| | - Kazuhiro Kobuke
- Howard Hughes Medical Institute, Departments of Molecular Physiology and Biophysics, Neurology, and Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Kevin P Campbell
- Howard Hughes Medical Institute, Departments of Molecular Physiology and Biophysics, Neurology, and Internal Medicine, The University of Iowa Roy J. and Lucille A. Carver College of Medicine, Iowa City, IA 52242 USA
| | - Michael Sinnreich
- Neuromuscular Research Group, Departments of Neurology and Biomedicine, University and University Hospital Basel, Petersgraben 4, 4031 Basel, Switzerland
| |
Collapse
|
21
|
Boisgérault F, Mingozzi F. The Skeletal Muscle Environment and Its Role in Immunity and Tolerance to AAV Vector-Mediated Gene Transfer. Curr Gene Ther 2015; 15:381-94. [PMID: 26122097 PMCID: PMC4515578 DOI: 10.2174/1566523215666150630121750] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Revised: 06/15/2015] [Accepted: 06/19/2015] [Indexed: 02/08/2023]
Abstract
Since the early days of gene therapy, muscle has been one the most studied tissue targets for the correction of enzyme deficiencies and myopathies. Several preclinical and clinical studies have been conducted using adeno-associated virus (AAV) vectors. Exciting progress has been made in the gene delivery technologies, from the identification of novel AAV serotypes to the development of novel vector delivery techniques. In parallel, significant knowledge has been generated on the host immune system and its interaction with both the vector and the transgene at the muscle level. In particular, the role of underlying muscle inflammation, characteristic of several diseases affecting the muscle, has been defined in terms of its potential detrimental impact on gene transfer with AAV vectors. At the same time, feedback immunomodulatory mechanisms peculiar of skeletal muscle involving resident regulatory T cells have been identified, which seem to play an important role in maintaining, at least to some extent, muscle homeostasis during inflammation and regenerative processes. Devising strategies to tip this balance towards unresponsiveness may represent an avenue to improve the safety and efficacy of muscle gene transfer with AAV vectors.
Collapse
Affiliation(s)
| | - Federico Mingozzi
- Genethon, Evry, France
- University Pierre and Marie Curie, Paris, France
| |
Collapse
|
22
|
A dystrophic muscle broadens the contribution and activation of immune cells reacting to rAAV gene transfer. Gene Ther 2014; 21:828-39. [PMID: 25030611 PMCID: PMC4283385 DOI: 10.1038/gt.2014.61] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 04/16/2014] [Accepted: 05/20/2014] [Indexed: 01/12/2023]
Abstract
Recombinant adeno-associated viral vectors (rAAVs) are used for therapeutic gene transfer in skeletal muscle, but it is unclear if immune reactivity to gene transfer and persistence of transgene are affected by pathologic conditions such as muscular dystrophy. Thus, we compared dystrophic mice devoid of α-sarcoglycan with healthy mice to characterize immune cell activation and cellular populations contributing to the loss of gene-modified myofibers. Following rAAV2/1 delivery of an immunogenic α-sarcoglycan reporter transgene in the muscle, both strains developed strong CD4 and CD8 T-cell-mediated immune responses in lymphoid organs associated with muscle CD3+ T and CD11b+ mononuclear cell infiltrates. Selective cell subset depletion models revealed that CD4+ T cells were essential for transgene rejection in both healthy and pathologic mice, but macrophages and CD8+ T cells additionally contributed as effector cells of transgene rejection only in dystrophic mice. Vectors restricting transgene expression in antigen-presenting cells showed that endogenous presentation of transgene products was the sole mechanism responsible for T-cell priming in normal mice, whereas additional and protracted antigenic presentation occurred in dystrophic animals, leading to secondary CD4+ T-cell activation and failure to maintain transgene expression. Therefore, the dystrophic environment diversifies cellular immune response mechanisms induced by gene transfer, with a negative outcome.
Collapse
|
23
|
de Oliveira BM, Matsumura CY, Fontes-Oliveira CC, Gawlik KI, Acosta H, Wernhoff P, Durbeej M. Quantitative proteomic analysis reveals metabolic alterations, calcium dysregulation, and increased expression of extracellular matrix proteins in laminin α2 chain-deficient muscle. Mol Cell Proteomics 2014; 13:3001-13. [PMID: 24994560 DOI: 10.1074/mcp.m113.032276] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Congenital muscular dystrophy with laminin α2 chain deficiency (MDC1A) is one of the most severe forms of muscular disease and is characterized by severe muscle weakness and delayed motor milestones. The genetic basis of MDC1A is well known, yet the secondary mechanisms ultimately leading to muscle degeneration and subsequent connective tissue infiltration are not fully understood. In order to obtain new insights into the molecular mechanisms underlying MDC1A, we performed a comparative proteomic analysis of affected muscles (diaphragm and gastrocnemius) from laminin α2 chain-deficient dy(3K)/dy(3K) mice, using multidimensional protein identification technology combined with tandem mass tags. Out of the approximately 700 identified proteins, 113 and 101 proteins, respectively, were differentially expressed in the diseased gastrocnemius and diaphragm muscles compared with normal muscles. A large portion of these proteins are involved in different metabolic processes, bind calcium, or are expressed in the extracellular matrix. Our findings suggest that metabolic alterations and calcium dysregulation could be novel mechanisms that underlie MDC1A and might be targets that should be explored for therapy. Also, detailed knowledge of the composition of fibrotic tissue, rich in extracellular matrix proteins, in laminin α2 chain-deficient muscle might help in the design of future anti-fibrotic treatments. All MS data have been deposited in the ProteomeXchange with identifier PXD000978 (http://proteomecentral.proteomexchange.org/dataset/PXD000978).
Collapse
Affiliation(s)
- Bruno Menezes de Oliveira
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Cintia Y Matsumura
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden; ¶Departament of Functional and Structural Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | - Cibely C Fontes-Oliveira
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Kinga I Gawlik
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Helena Acosta
- ‖Stem Cell Center, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Patrik Wernhoff
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden
| | - Madeleine Durbeej
- From the §Department of Experimental Medical Science, Unit of Muscle Biology, Lund University, BMC B12, 221 84 Lund, Sweden;
| |
Collapse
|
24
|
Barfield WL, Uaesoontrachoon K, Wu CS, Lin S, Chen Y, Wang PC, Kanaan Y, Bond V, Hoffman EP. Eccentric muscle challenge shows osteopontin polymorphism modulation of muscle damage. Hum Mol Genet 2014; 23:4043-50. [PMID: 24626632 PMCID: PMC4082368 DOI: 10.1093/hmg/ddu118] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
A promoter polymorphism of the osteopontin (OPN) gene (rs28357094) has been associated with multiple inflammatory states, severity of Duchenne muscular dystrophy (DMD) and muscle size in healthy young adults. We sought to define the mechanism of action of the polymorphism, using allele-specific in vitro reporter assays in muscle cells, and a genotype-stratified intervention in healthy controls. In vitro reporter constructs showed the G allele to respond to estrogen treatment, whereas the T allele showed no transcriptional response. Young adult volunteers (n = 187) were enrolled into a baseline study, and subjects with specific rs28357094 genotypes enrolled into an eccentric muscle challenge intervention [n = 3 TT; n = 3 GG/GT (dominant inheritance model)]. Female volunteers carrying the G allele showed significantly greater inflammation and increased muscle volume change as determined by magnetic resonance imaging T1- and T2-weighted images after eccentric challenge, as well as greater decrement in biceps muscle force. Our data suggest a model where the G allele enables enhanced activities of upstream enhancer elements due to loss of Sp1 binding at the polymorphic site. This results in significantly greater expression of the pro-inflammatory OPN cytokine during tissue remodeling in response to challenge in G allele carriers, promoting muscle hypertrophy in normal females, but increased damage in DMD patients.
Collapse
Affiliation(s)
| | | | - Chung-Sheih Wu
- Department of Radiology, Howard University School of Medicine, Washington, DC, USA
| | - Stephen Lin
- Department of Radiology, Howard University School of Medicine, Washington, DC, USA
| | - Yue Chen
- Department of Radiology, Howard University School of Medicine, Washington, DC, USA
| | - Paul C Wang
- Department of Radiology, Howard University School of Medicine, Washington, DC, USA
| | | | - Vernon Bond
- Department of Health, Human Performance and Leisure Studies, Howard University, Washington, DC, USA
| | - Eric P Hoffman
- Center for Genetic Medicine Research, Children's National Medical Center, Washington, DC, USA
| |
Collapse
|
25
|
Kerr JP, Ward CW, Bloch RJ. Dysferlin at transverse tubules regulates Ca(2+) homeostasis in skeletal muscle. Front Physiol 2014; 5:89. [PMID: 24639655 PMCID: PMC3944681 DOI: 10.3389/fphys.2014.00089] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Accepted: 02/15/2014] [Indexed: 11/13/2022] Open
Abstract
The class of muscular dystrophies linked to the genetic ablation or mutation of dysferlin, including Limb Girdle Muscular Dystrophy 2B (LGMD2B) and Miyoshi Myopathy (MM), are late-onset degenerative diseases. In lieu of a genetic cure, treatments to prevent or slow the progression of dysferlinopathy are of the utmost importance. Recent advances in the study of dysferlinopathy have highlighted the necessity for the maintenance of calcium handling in altering or slowing the progression of muscular degeneration resulting from the loss of dysferlin. This review highlights new evidence for a role for dysferlin at the transverse (t-) tubule of striated muscle, where it is involved in maintaining t-tubule structure and function.
Collapse
Affiliation(s)
- Jaclyn P Kerr
- Department of Physiology, University of Maryland School of Medicine Baltimore, MD, USA
| | - Christopher W Ward
- Department of Organizational Systems and Adult Health, University of Maryland School of Nursing Baltimore, MD, USA
| | - Robert J Bloch
- Department of Physiology, University of Maryland School of Medicine Baltimore, MD, USA
| |
Collapse
|
26
|
Kombairaju P, Kerr JP, Roche JA, Pratt SJP, Lovering RM, Sussan TE, Kim JH, Shi G, Biswal S, Ward CW. Genetic silencing of Nrf2 enhances X-ROS in dysferlin-deficient muscle. Front Physiol 2014; 5:57. [PMID: 24600403 PMCID: PMC3928547 DOI: 10.3389/fphys.2014.00057] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 01/29/2014] [Indexed: 11/13/2022] Open
Abstract
Oxidative stress is a critical disease modifier in the muscular dystrophies. Recently, we discovered a pathway by which mechanical stretch activates NADPH Oxidase 2 (Nox2) dependent ROS generation (X-ROS). Our work in dystrophic skeletal muscle revealed that X-ROS is excessive in dystrophin-deficient (mdx) skeletal muscle and contributes to muscle injury susceptibility, a hallmark of the dystrophic process. We also observed widespread alterations in the expression of genes associated with the X-ROS pathway and redox homeostasis in muscles from both Duchenne muscular dystrophy patients and mdx mice. As nuclear factor erythroid 2-related factor 2 (Nrf2) plays an essential role in the transcriptional regulation of genes involved in redox homeostasis, we hypothesized that Nrf2 deficiency may contribute to enhanced X-ROS signaling by reducing redox buffering. To directly test the effect of diminished Nrf2 activity, Nrf2 was genetically silenced in the A/J model of dysferlinopathy—a model with a mild histopathologic and functional phenotype. Nrf2-deficient A/J mice exhibited significant muscle-specific functional deficits, histopathologic abnormalities, and dramatically enhanced X-ROS compared to control A/J and WT mice, both with functional Nrf2. Having identified that reduced Nrf2 activity is a negative disease modifier, we propose that strategies targeting Nrf2 activation may address the generalized reduction in redox homeostasis to halt or slow dystrophic progression.
Collapse
Affiliation(s)
- Ponvijay Kombairaju
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University Baltimore, MD, USA
| | - Jaclyn P Kerr
- Department of Physiology, University of Maryland School of Medicine Baltimore, MD, USA
| | - Joseph A Roche
- Department of Physiology, University of Maryland School of Medicine Baltimore, MD, USA
| | - Stephen J P Pratt
- Department of Orthopaedics, University of Maryland School of Medicine Baltimore, MD, USA
| | - Richard M Lovering
- Department of Orthopaedics, University of Maryland School of Medicine Baltimore, MD, USA
| | - Thomas E Sussan
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University Baltimore, MD, USA
| | - Jung-Hyun Kim
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University Baltimore, MD, USA
| | - Guoli Shi
- Department of Organizational Systems and Adult Health, University of Maryland School of Nursing Baltimore, MD, USA
| | - Shyam Biswal
- Department of Environmental Health Sciences, Bloomberg School of Public Health, Johns Hopkins University Baltimore, MD, USA
| | - Christopher W Ward
- Department of Organizational Systems and Adult Health, University of Maryland School of Nursing Baltimore, MD, USA
| |
Collapse
|
27
|
Boisgerault F, Gross DA, Ferrand M, Poupiot J, Darocha S, Richard I, Galy A. Prolonged gene expression in muscle is achieved without active immune tolerance using microrRNA 142.3p-regulated rAAV gene transfer. Hum Gene Ther 2014; 24:393-405. [PMID: 23427817 DOI: 10.1089/hum.2012.208] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Gene transfer efficacy is limited by unwanted immunization against transgene products. In some models, immunization may be avoided by regulating transgene expression with mir142.3p target sequences. Yet, it is unclear if such a strategy controls T-cell responses following recombinant adeno-associated viral vector (rAAV)-mediated gene transfer, particularly in muscle. In mice, intramuscular rAAV1 gene delivery of a tagged human sarcoglycan muscle protein is robustly immunogenic and leads to muscle destruction. In this model, the simple insertion of mir142.3p-target sequences in the transgene expression cassette modifies the outcome of gene transfer, providing high and persistent levels of muscle transduction in C57Bl/6 mice. Such regulated vector fails to prime specific CD4 and CD8 T cells; although, transgene tolerance seems to result from ignorance and could be broken by a robust antigenic challenge. While effective in normal mice, the mir142.3p-regulated transgene remains immunogenic in sarcoglycan-deficient dystrophic mice. In these mice, transgene expression is only prolonged but does not persist as effector CD4 and CD8 T-cell responses develop. Thus, using a mir142.3p-regulated transgene can improve rAAV muscle gene transfer results, but the level of efficacy depends on the context of application. In normal muscle, this strategy is sufficient to prevent immunization and functions even more effectively than tissue-specific promoters. In dystrophic models, additional strategies are required to fully control T-cell responses.
Collapse
Affiliation(s)
- Florence Boisgerault
- Genethon, Molecular Immunology and Innovative Biotherapies Group, Evry F91002 France
| | | | | | | | | | | | | |
Collapse
|
28
|
Townsend D, Yasuda S, Metzger J. Cardiomyopathy of Duchenne muscular dystrophy: pathogenesis and prospect of membrane sealants as a new therapeutic approach. Expert Rev Cardiovasc Ther 2014; 5:99-109. [PMID: 17187461 DOI: 10.1586/14779072.5.1.99] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a devastating progressive disease of striated muscle deterioration. This fatal X-linked disorder results from the loss of the protein dystrophin, which in turn causes striated muscle membrane instability. Cardiac dysfunction is a growing problem in patients with DMD, but relatively little is known about the pathophysiology of the dystrophic heart. At present, there is no effective treatment for DMD and the current clinical approaches are primarily supportive in nature. This review will discuss the pathogenesis of DMD in the heart and discuss how these pathogenic processes have led to a new class of agents directed specifically at restoring membrane integrity to dystrophic myocardium. The tri-block poloxamers, specifically poloxamer 188 (P188), are able to stabilize the membranes of dystrophic myocardium in animal models and may offer a new therapeutic approach for cardiac disease in DMD.
Collapse
Affiliation(s)
- DeWayne Townsend
- Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, Michigan, USA.
| | | | | |
Collapse
|
29
|
Abstract
Neuromuscular diseases, which encompass disorders that affect muscle and its innervation, are highly heritable. Genetic diagnosis now frequently pinpoints the primary mutation responsible for a given neuromuscular disease. However, the results from genetic testing indicate that neuromuscular disease phenotypes may vary widely, even in individuals with the same primary disease-causing mutation. Clinical variability arises from both genetic and environmental factors. Genetic modifiers can now be identified using candidate gene as well as genomic approaches. The presence of genetic modifiers for neuromuscular disease helps define the clinical outcome and also highlights pathways of potential therapeutic utility. Herein, we will focus on single gene neuromuscular disorders, including muscular dystrophy, spinal muscular atrophy, and amyotrophic lateral sclerosis, and the methods that have been used to identify modifier genes. Animal models have been an invaluable resource for modifier gene discovery and subsequent mechanistic studies. Some modifiers, identified using animal models, have successfully translated to the human counterpart. Furthermore, in a few instances, modifier gene discovery has repetitively uncovered the same pathway, such as TGFβ signaling in muscular dystrophy, further emphasizing the relevance of that pathway. Knowledge of genetic factors that influence disease can have direct clinical applications for prognosis and predicted outcome.
Collapse
Affiliation(s)
- Kay-Marie Lamar
- Department of Human Genetics, Department of Medicine, Section of Cardiology, The University of Chicago, Chicago, IL, USA
| | - Elizabeth M McNally
- Department of Human Genetics, Department of Medicine, Section of Cardiology, The University of Chicago, Chicago, IL, USA
| |
Collapse
|
30
|
Pagel CN, Wasgewatte Wijesinghe DK, Taghavi Esfandouni N, Mackie EJ. Osteopontin, inflammation and myogenesis: influencing regeneration, fibrosis and size of skeletal muscle. J Cell Commun Signal 2013; 8:95-103. [PMID: 24318932 DOI: 10.1007/s12079-013-0217-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Accepted: 11/25/2013] [Indexed: 12/20/2022] Open
Abstract
Osteopontin is a multifunctional matricellular protein that is expressed by many cell types. Through cell-matrix and cell-cell interactions the molecule elicits a number of responses from a broad range of target cells via its interaction with integrins and the hyaluronan receptor CD44. In many tissues osteopontin has been found to be involved in important physiological and pathological processes, including tissue repair, inflammation and fibrosis. Post-natal skeletal muscle is a highly differentiated and specialised tissue that retains a remarkable capacity for regeneration following injury. Regeneration of skeletal muscle requires the co-ordinated activity of inflammatory cells that infiltrate injured muscle and are responsible for initiating muscle fibre degeneration and phagocytosis of necrotic tissue, and muscle precursor cells that regenerate the injured muscle fibres. This review focuses on the current evidence that osteopontin plays multiple roles in skeletal muscle, with particular emphasis on its role in regeneration and fibrosis following injury, and in determining the severity of myopathic diseases such as Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Charles N Pagel
- Faculty of Veterinary Science, University of Melbourne, Parkville, Victoria, 3010, Australia,
| | | | | | | |
Collapse
|
31
|
Krajacic P, Pistilli EE, Tanis JE, Khurana TS, Lamitina ST. FER-1/Dysferlin promotes cholinergic signaling at the neuromuscular junction in C. elegans and mice. Biol Open 2013; 2:1245-52. [PMID: 24244862 PMCID: PMC3828772 DOI: 10.1242/bio.20135637] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Accepted: 09/25/2013] [Indexed: 12/28/2022] Open
Abstract
Dysferlin is a member of the evolutionarily conserved ferlin gene family. Mutations in Dysferlin lead to Limb Girdle Muscular Dystrophy 2B (LGMD2B), an inherited, progressive and incurable muscle disorder. However, the molecular mechanisms underlying disease pathogenesis are not fully understood. We found that both loss-of-function mutations and muscle-specific overexpression of C. elegans fer-1, the founding member of the Dysferlin gene family, caused defects in muscle cholinergic signaling. To determine if Dysferlin-dependent regulation of cholinergic signaling is evolutionarily conserved, we examined the in vivo physiological properties of skeletal muscle synaptic signaling in a mouse model of Dysferlin-deficiency. In addition to a loss in muscle strength, Dysferlin −/− mice also exhibited a cholinergic deficit manifested by a progressive, frequency-dependent decrement in their compound muscle action potentials following repetitive nerve stimulation, which was observed in another Dysferlin mouse model but not in a Dysferlin-independent mouse model of muscular dystrophy. Oral administration of Pyridostigmine bromide, a clinically used acetylcholinesterase inhibitor (AchE.I) known to increase synaptic efficacy, reversed the action potential defect and restored in vivo muscle strength to Dysferlin −/− mice without altering muscle pathophysiology. Our data demonstrate a previously unappreciated role for Dysferlin in the regulation of cholinergic signaling and suggest that such regulation may play a significant pathophysiological role in LGMD2B disease.
Collapse
Affiliation(s)
- Predrag Krajacic
- Department of Physiology, Richards Research Building A702, University of Pennsylvania , Philadelphia, PA 19104 , USA ; Pennsylvania Muscle Institute, 700A Clinical Research Building, University of Pennsylvania , Philadelphia, PA 19104 , USA
| | | | | | | | | |
Collapse
|
32
|
Intrinsic epigenetic regulation of the D4Z4 macrosatellite repeat in a transgenic mouse model for FSHD. PLoS Genet 2013; 9:e1003415. [PMID: 23593020 PMCID: PMC3616921 DOI: 10.1371/journal.pgen.1003415] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2012] [Accepted: 02/11/2013] [Indexed: 11/19/2022] Open
Abstract
Facioscapulohumeral dystrophy (FSHD) is a progressive muscular dystrophy caused by decreased epigenetic repression of the D4Z4 macrosatellite repeats and ectopic expression of DUX4, a retrogene encoding a germline transcription factor encoded in each repeat. Unaffected individuals generally have more than 10 repeats arrayed in the subtelomeric region of chromosome 4, whereas the most common form of FSHD (FSHD1) is caused by a contraction of the array to fewer than 10 repeats, associated with decreased epigenetic repression and variegated expression of DUX4 in skeletal muscle. We have generated transgenic mice carrying D4Z4 arrays from an FSHD1 allele and from a control allele. These mice recapitulate important epigenetic and DUX4 expression attributes seen in patients and controls, respectively, including high DUX4 expression levels in the germline, (incomplete) epigenetic repression in somatic tissue, and FSHD–specific variegated DUX4 expression in sporadic muscle nuclei associated with D4Z4 chromatin relaxation. In addition we show that DUX4 is able to activate similar functional gene groups in mouse muscle cells as it does in human muscle cells. These transgenic mice therefore represent a valuable animal model for FSHD and will be a useful resource to study the molecular mechanisms underlying FSHD and to test new therapeutic intervention strategies. Facioscapulohumeral dystrophy (FSHD) is a progressive muscle disorder that is associated with contraction and chromatin relaxation of the D4Z4 macrosatellite repeat on chromosome 4q. Each unit of the repeat contains a copy of the primate-specific DUX4 retrogene, encoding a germline transcription factor that is repressed in somatic tissue. In FSHD, somatic repression of the DUX4 gene is compromised, leading to a variegated expression pattern of DUX4 in muscle cells. The complex (epi)genetic etiology of FSHD has long hampered the generation of a faithful animal model, and thus far the role of FSHD candidate genes has only been studied in model organisms by overexpression approaches. Here we present two transgenic mouse models containing either patient- or control-sized D4Z4 repeats. In our mice, the regulation of the FSHD locus is preserved in both lines, and only in the disease model somatic derepression and variegated expression of DUX4 is observed. These mice thus reflect many aspects of the complex regulation of DUX4 expression in humans. These models may therefore become valuable tools in understanding the in vivo regulation and function of DUX4, its role in FSHD, and the evaluation of therapeutic strategies.
Collapse
|
33
|
Marshall JL, Crosbie-Watson RH. Sarcospan: a small protein with large potential for Duchenne muscular dystrophy. Skelet Muscle 2013; 3:1. [PMID: 23282144 PMCID: PMC3599653 DOI: 10.1186/2044-5040-3-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2012] [Accepted: 11/27/2012] [Indexed: 01/09/2023] Open
Abstract
Purification of the proteins associated with dystrophin, the gene product responsible for Duchenne muscular dystrophy, led to the discovery of the dystrophin-glycoprotein complex. Sarcospan, a 25-kDa transmembrane protein, was the last component to be identified and its function in skeletal muscle has been elusive. This review will focus on progress over the last decade revealing that sarcospan is an important regulator of muscle cell adhesion, strength, and regeneration. Investigations using several transgenic mouse models demonstrate that overexpression of sarcospan in the mouse model for Duchenne muscular dystrophy ameliorates pathology and restores muscle cell binding to laminin. Sarcospan improves cell surface expression of the dystrophin- and utrophin-glycoprotein complexes as well as α7β1 integrin, which are the three major laminin-binding complexes in muscle. Utrophin and α7β1 integrin compensate for the loss of dystrophin and the finding that sarcospan increases their abundance at the extra-synaptic sarcolemma supports the use of sarcospan as a therapeutic target. Newly discovered phenotypes in sarcospan-deficient mice, including a reduction in specific force output and increased drop in force in the diaphragm muscle, result from decreased utrophin and dystrophin expression and further reveal sarcospan’s role in determining abundance of these complexes. Dystrophin protein levels and the specific force output of the diaphragm muscle are further reduced upon genetic removal of α7 integrin (Itga7) in SSPN-deficient mice, demonstrating that interactions between integrin and sarcospan are critical for maintenance of the dystrophin-glycoprotein complex and force production of the diaphragm muscle. Sarcospan is a major regulator of Akt signaling pathways and sarcospan-deficiency significantly impairs muscle regeneration, a process that is dependent on Akt activation. Intriguingly, sarcospan regulates glycosylation of a specific subpopulation of α-dystroglycan, the laminin-binding receptor associated with dystrophin and utrophin, localized to the neuromuscular junction. Understanding the basic mechanisms responsible for assembly and trafficking of the dystrophin- and utrophin-glycoprotein complexes to the cell surface is lacking and recent studies suggest that sarcospan plays a role in these essential processes.
Collapse
Affiliation(s)
- Jamie L Marshall
- Department of Integrative Biology and Physiology, University of California Los Angeles, 610 Charles E, Young Drive East, Terasaki Life Sciences Building, Los Angeles, CA, 90095, USA.
| | | |
Collapse
|
34
|
Garbe CS, Buttgereit A, Schurmann S, Friedrich O. Automated Multiscale Morphometry of Muscle Disease From Second Harmonic Generation Microscopy Using Tensor-Based Image Processing. IEEE Trans Biomed Eng 2012; 59:39-44. [DOI: 10.1109/tbme.2011.2167325] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
35
|
Belfiore MP, Berritto D, Iacobellis F, Rossi C, Nigro G, Rotundo IL, Cozzolino S, Cappabianca S, Rotondo A, Grassi R. A longitudinal study on BIO14.6 hamsters with dilated cardiomyopathy: micro-echocardiographic evaluation. Cardiovasc Ultrasound 2011; 9:39. [PMID: 22151912 PMCID: PMC3254069 DOI: 10.1186/1476-7120-9-39] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2011] [Accepted: 12/08/2011] [Indexed: 11/23/2022] Open
Abstract
Background In recent years, several new technologies for small-animal imaging have been developed. In particular, the use of ultrasound in animal imaging has focused on the investigation of accessible biological structures such as the heart, of which it provides a morphological and functional assessment. The purpose of this study was to investigate the role of micro-ultrasonography (μ-US) in a longitudinal study on BIO14.6 cardiomyopathic hamsters treated with gene therapy. Methods Thirty hamsters were divided into three groups (n = 10): Group I, untreated BIO 14.6 hamsters; Group II, BIO 14.6 hamsters treated with gene therapy; Group III, untreated wild type (WT) hamsters. All hamsters underwent serial μ-US sessions and were sacrificed at predetermined time points. Results μ-US revealed: in Group I, progressive dilation of the left ventricle with a change in heart morphology from an elliptical to a more spherical shape, altered configuration of the mitral valve and subvalvular apparatus, and severe reduction in ejection fraction; in Group II, mild decrease in contractile function and ejection fraction; in Group III, normal cardiac chamber morphology and function. There was a negative correlation between the percentage of fibrosis observed at histology and the ejection fraction obtained on μ-echocardiography (Spearman r: -0.839; p < 0.001). Conclusions Although histological examination remains indispensable for a conclusive diagnosis, high-frequency μ-echocardiography, thanks to the high spatial and contrast resolution, can be considered sufficient for monitoring therapeutic efficacy and/or the progression of dilated cardiomyopathy, providing an alternative tool for repeatable and noninvasive evaluation.
Collapse
Affiliation(s)
- Maria Paola Belfiore
- Institute of Radiology, Second University of Naples, P zza Miraglia 2, 80138 Napoli, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Serum matrix metalloproteinase-9 (MMP-9) as a biomarker for monitoring disease progression in Duchenne muscular dystrophy (DMD). Neuromuscul Disord 2011; 21:569-78. [DOI: 10.1016/j.nmd.2011.05.011] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 05/24/2011] [Accepted: 05/27/2011] [Indexed: 11/19/2022]
|
37
|
Goonasekera SA, Lam CK, Millay DP, Sargent MA, Hajjar RJ, Kranias EG, Molkentin JD. Mitigation of muscular dystrophy in mice by SERCA overexpression in skeletal muscle. J Clin Invest 2011; 121:1044-52. [PMID: 21285509 DOI: 10.1172/jci43844] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 12/01/2010] [Indexed: 11/17/2022] Open
Abstract
Muscular dystrophies (MDs) comprise a group of degenerative muscle disorders characterized by progressive muscle wasting and often premature death. The primary defect common to most MDs involves disruption of the dystrophin-glycoprotein complex (DGC). This leads to sarcolemmal instability and Ca(2+) influx, inducing cellular necrosis. Here we have shown that the dystrophic phenotype observed in δ-sarcoglycan–null (Sgcd(–/–)) mice and dystrophin mutant mdx mice is dramatically improved by skeletal muscle–specific overexpression of sarcoplasmic reticulum Ca(2+) ATPase 1 (SERCA1). Rates of myofiber central nucleation, tissue fibrosis, and serum creatine kinase levels were dramatically reduced in Sgcd(–/–) and mdx mice with the SERCA1 transgene, which also rescued the loss of exercise capacity in Sgcd(–/–) mice. Adeno-associated virus–SERCA2a (AAV-SERCA2a) gene therapy in the gastrocnemius muscle of Sgcd(–/–) mice mitigated dystrophic disease. SERCA1 overexpression reversed a defect in sarcoplasmic reticulum Ca(2+) reuptake that characterizes dystrophic myofibers and reduced total cytosolic Ca(2+). Further, SERCA1 overexpression almost completely rescued the dystrophic phenotype in a mouse model of MD driven solely by Ca(2+) influx. Mitochondria isolated from the muscle of SERCA1-Sgcd(–/–) mice were no longer swollen and calpain activation was reduced, suggesting protection from Ca(2+)-driven necrosis. Our results suggest a novel therapeutic approach using SERCA1 to abrogate the altered intracellular Ca(2+) levels that underlie most forms of MD.
Collapse
Affiliation(s)
- Sanjeewa A Goonasekera
- Department of Pediatrics, University of Cincinnati, Cincinnati Children's Hospital Medical Center, Howard Hughes Medical Institute, Cincinnati, Ohio 45229, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Kobayashi K, Izawa T, Kuwamura M, Yamate J. Comparative Gene Expression Analysis in the Skeletal Muscles of Dysferlin-deficient SJL/J and A/J Mice. J Toxicol Pathol 2011; 24:49-62. [PMID: 22272044 PMCID: PMC3234620 DOI: 10.1293/tox.24.49] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2010] [Accepted: 12/01/2010] [Indexed: 11/23/2022] Open
Abstract
Quantitative real-time polymerase chain reaction (qRT-PCR) analysis was conducted to
determine whether or not there are interstrain or site-dependent differences in the gene
expression profiles of skeletal muscles in SJL/J and A/J mice as dysferlinopathy models.
Upon analysis by qRT-PCR, SJL/J mice showed a trend of increased gene expression level of
uncoupling protein 2 in the rectus femoris and longissimus lumborum at 30 weeks of age
when dystrophic lesions became histopathologically pronounced. Heme oxygenase 1 and S100
calcium binding protein A4 were upregulated in the rectus femoris, longissimus lumborum
and abdominal muscles, in which dystrophic lesions occur more commonly in SJL mice. The
gene expression levels of heat shock protein 70 in most muscles of A/J mice were lower
than those of BALB/c mice as control. SJL/J mice exhibited a marked lowering of
decay-accelerating factor 1/CD55 gene expression level in all studied muscles except for
the heart at all ages compared with that of BALB/c mice. This study showed that there were
some interstrain differences in the gene expres sion profiles of skeletal muscles between
SJL/J and A/J mice. Further investigation is required to reveal whether these alterations
of the expression levels are the cause of dystrophic changes or occur subsequent to muscle
damage.
Collapse
Affiliation(s)
- Kinji Kobayashi
- Laboratory of Veterinary Pathology, Division of Veterinary Sciences, Graduate School of Life and Environmental Sciences, Osaka Prefecture University, 1-58 Rinku-ourai-kita, Izumisano City, Osaka 598-8531, Japan
| | | | | | | |
Collapse
|
39
|
Pegoraro E, Hoffman EP, Piva L, Gavassini BF, Cagnin S, Ermani M, Bello L, Soraru G, Pacchioni B, Bonifati MD, Lanfranchi G, Angelini C, Kesari A, Lee I, Gordish-Dressman H, Devaney JM, McDonald CM. SPP1 genotype is a determinant of disease severity in Duchenne muscular dystrophy. Neurology 2010; 76:219-26. [PMID: 21178099 DOI: 10.1212/wnl.0b013e318207afeb] [Citation(s) in RCA: 169] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
OBJECTIVE Duchenne muscular dystrophy (DMD) is the most common single-gene lethal disorder. Substantial patient-patient variability in disease onset and progression and response to glucocorticoids is seen, suggesting genetic or environmental modifiers. METHODS Two DMD cohorts were used as test and validation groups to define genetic modifiers: a Padova longitudinal cohort (n = 106) and the Cooperative International Neuromuscular Research Group (CINRG) cross-sectional natural history cohort (n = 156). Single nucleotide polymorphisms to be genotyped were selected from mRNA profiling in patients with severe vs mild DMD, and genome-wide association studies in metabolism and polymorphisms influencing muscle phenotypes in normal volunteers were studied. RESULTS Effects on both disease progression and response to glucocorticoids were observed with polymorphism rs28357094 in the gene promoter of SPP1 (osteopontin). The G allele (dominant model; 35% of subjects) was associated with more rapid progression (Padova cohort log rank p = 0.003), and 12%-19% less grip strength (CINRG cohort p = 0.0003). CONCLUSIONS Osteopontin genotype is a genetic modifier of disease severity in Duchenne dystrophy. Inclusion of genotype data as a covariate or in inclusion criteria in DMD clinical trials would reduce intersubject variance, and increase sensitivity of the trials, particularly in older subjects.
Collapse
Affiliation(s)
- E Pegoraro
- Neuromuscular Center, Department of Neurosciences, University of Padova, 35128 Padova, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Frongillo EA, Isaacman TD, Horan CM, Wethington E, Pillemer K. Adequacy of and satisfaction with delivery and use of home-delivered meals. ACTA ACUST UNITED AC 2010; 29:211-26. [PMID: 20473813 DOI: 10.1080/01639361003772525] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
For home-delivered meals to have a beneficial impact on older persons, it is important that both delivery of services and use by older persons are adequate. From November 2004 to February 2005, we conducted a random-sample telephone survey of 1505 New York City home-delivered meals recipients, asking them about adequacy of and satisfaction with delivery of services and use of meal services. Fourteen percent of recipients relied solely on program food. Two-thirds prepared other foods themselves. Consumption of fruit, vegetables, and milk was low; 14-20% of recipients consumed each of these less than 1 time per day. Most recipients saw (and about half talked with) the meal deliverer most of the time. Most could contact the meal provider agency, but had not done so. A second stratified sample of 500 meal recipients was surveyed in June 2006 regarding satisfaction with food packaging and labels, food acquisition, meal delivery, and meal variety. About three-fourths of recipients reported satisfaction most of the time with the meals in terms of taste, variety, ease of preparation, healthfulness, and fit to religious or cultural needs. The most satisfied recipients were those who were receiving hot meals, food-secure, without hearing problems, frailer, in better emotional health, with informal social support, and more religious.
Collapse
Affiliation(s)
- Edward A Frongillo
- Department of Health Promotion, Education, and Behavior, University of South Carolina, 800 Sumter St., Room 216, Columbia, SC 29208, USA.
| | | | | | | | | |
Collapse
|
41
|
Hestand MS, Klingenhoff A, Scherf M, Ariyurek Y, Ramos Y, van Workum W, Suzuki M, Werner T, van Ommen GJB, den Dunnen JT, Harbers M, 't Hoen PAC. Tissue-specific transcript annotation and expression profiling with complementary next-generation sequencing technologies. Nucleic Acids Res 2010; 38:e165. [PMID: 20615900 PMCID: PMC2938216 DOI: 10.1093/nar/gkq602] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Next-generation sequencing is excellently suited to evaluate the abundance of mRNAs to study gene expression. Here we compare two alternative technologies, cap analysis of gene expression (CAGE) and serial analysis of gene expression (SAGE), for the same RNA samples. Along with quantifying gene expression levels, CAGE can be used to identify tissue-specific transcription start sites, while SAGE monitors 3′-end usage. We used both methods to get more insight into the transcriptional control of myogenesis, studying differential gene expression in differentiated and proliferating C2C12 myoblast cells with statistical evaluation of reproducibility and differential gene expression. Both CAGE and SAGE provided highly reproducible data (Pearson's correlations >0.92 among biological triplicates). With both methods we found around 10 000 genes expressed at levels 2 transcripts per million (0.3 copies per cell), with an overlap of 86%. We identified 4304 and 3846 genes differentially expressed between proliferating and differentiated C2C12 cells by CAGE and SAGE, respectively, with an overlap of 2144. We identified 196 novel regulatory regions with preferential use in proliferating or differentiated cells. Next-generation sequencing of CAGE and SAGE libraries provides consistent expression levels and can enrich current genome annotations with tissue-specific promoters and alternative 3′-UTR usage.
Collapse
Affiliation(s)
- Matthew S Hestand
- The Center for Human and Clinical Genetics, Leiden University Medical Center, 2300 RC Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Soluble TNF-α receptor secretion from healthy or dystrophic mice after AAV6-mediated muscle gene transfer. Gene Ther 2010; 17:1400-10. [PMID: 20596058 DOI: 10.1038/gt.2010.94] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Muscle is an attractive target because it is easily accessible; it also offers a permissive environment for adeno-associated virus (AAV)-mediated gene transfer and has an abundant blood vascular supply providing an efficient transport system for the secretion of proteins. However, gene therapy of dystrophic muscle may be more difficult than that of healthy tissue because of degenerative-regenerative processes, and also because of the inflammatory context. In this study we followed the expression levels of secreted inhibitors of the proinflammatory tumor necrosis factor (TNF) cytokine after intramuscular (i.m.) injection of AAV6 into dystrophic mdx and healthy C57BL/10 mice. We used two chimeric proteins, namely, the human or murine TNF-soluble receptor I fused with the murine heavy immunoglobulin chain. We conducted an AAV6 dose-response study and determined the kinetics of transgene expression. In addition, we followed the antibody response against the transgenes and studied their expression pattern in the muscle. Our results show that transduction efficiency is reduced in dystrophic muscles as compared with healthy ones. Furthermore, we found that the immune response against the secreted protein is stronger in mdx mice. Together, our results underscore that the pathological state of the muscle has to be taken into consideration when designing gene therapy approaches.
Collapse
|
43
|
Qureshi MM, McClure WC, Arevalo NL, Rabon RE, Mohr B, Bose SK, McCord JM, Tseng BS. The Dietary Supplement Protandim Decreases Plasma Osteopontin and Improves Markers of Oxidative Stress in Muscular Dystrophy Mdx Mice. J Diet Suppl 2010; 7:159-178. [PMID: 20740052 DOI: 10.3109/19390211.2010.482041] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Therapeutic options for Duchenne muscular dystrophy (DMD), the most common and lethal neuromuscular disorder in children, remain elusive. Oxidative damage is implicated as a pertinent factor involved in its pathogenesis. Protandim((R)) is an over-the-counter supplement with the ability to induce antioxidant enzymes. In this study we investigated whether Protandim((R)) provided benefit using surrogate markers and functional measures in the dystrophin-deficient (mdx)mouse model of DMD. Male 3-week-old mdx mice were randomized into two treatment groups: control (receiving standard rodent chow) and Protandim((R))-supplemented standard rodent chow. The diets were continued for 6-week and 6-month studies. The endpoints included the oxidative stress marker thiobarbituric acid-reactive substances (TBARS), plasma osteopontin (OPN), plasma paraoxonase (PON1) activity, H&E histology, gadolinium-enhanced magnetic resonance imaging (MRI) of leg muscle and motor functional measurements. The Protandim((R)) chow diet in mdx mice for 6 months was safe and well tolerated. After 6 months of Protandim((R)), a 48% average decrease in plasma TBARS was seen; 0.92 nmol/mg protein in controls versus 0.48 nmol/mg protein in the Protandim((R)) group (p = .006). At 6 months, plasma OPN was decreased by 57% (p = .001) in the Protandim((R))-treated mice. Protandim((R)) increased the plasma antioxidant enzyme PON1 activity by 35% (p = .018). After 6 months, the mdx mice with Protandim((R)) showed 38% less MRI signal abnormality (p = .07) than mice on control diet. In this 6-month mdx mouse study, Protandim((R)) did not significantly alter motor function nor histological criteria.
Collapse
Affiliation(s)
- Muhammad Muddasir Qureshi
- Department of Pediatrics, Texas Tech University Health Sciences Center, Paul L. Foster School of Medicine, El Paso, TX. Earlier, he was associated with Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Friedrich O, Both M, Weber C, Schürmann S, Teichmann MDH, von Wegner F, Fink RHA, Vogel M, Chamberlain JS, Garbe C. Microarchitecture is severely compromised but motor protein function is preserved in dystrophic mdx skeletal muscle. Biophys J 2010; 98:606-16. [PMID: 20159157 DOI: 10.1016/j.bpj.2009.11.005] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2009] [Revised: 10/27/2009] [Accepted: 11/05/2009] [Indexed: 10/19/2022] Open
Abstract
Progressive force loss in Duchenne muscular dystrophy is characterized by degeneration/regeneration cycles and fibrosis. Disease progression may involve structural remodeling of muscle tissue. An effect on molecular motorprotein function may also be possible. We used second harmonic generation imaging to reveal vastly altered subcellular sarcomere microarchitecture in intact single dystrophic mdx muscle cells (approximately 1 year old). Myofibril tilting, twisting, and local axis deviations explain at least up to 20% of force drop during unsynchronized contractile activation as judged from cosine angle sums of myofibril orientations within mdx fibers. In contrast, in vitro motility assays showed unaltered sliding velocities of single mdx fiber myosin extracts. Closer quantification of the microarchitecture revealed that dystrophic fibers had significantly more Y-shaped sarcomere irregularities ("verniers") than wild-type fibers (approximately 130/1000 microm(3) vs. approximately 36/1000 microm(3)). In transgenic mini-dystrophin-expressing fibers, ultrastructure was restored (approximately 38/1000 microm(3) counts). We suggest that in aged dystrophic toe muscle, progressive force loss is reflected by a vastly deranged micromorphology that prevents a coordinated and aligned contraction. Second harmonic generation imaging may soon be available in routine clinical diagnostics, and in this work we provide valuable imaging tools to track and quantify ultrastructural worsening in Duchenne muscular dystrophy, and to judge the beneficial effects of possible drug or gene therapies.
Collapse
Affiliation(s)
- O Friedrich
- Medical Biophysics, Institute of Physiology and Pathophysiology, Ruprecht Karls University, Heidelberg, Germany.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Muscular dystrophies are individually rare genetic disorders that cause much chronic disability, affecting young children and adults. In the past 20 years, more than 30 genetic types of muscular dystrophy have been defined. During this time, precise diagnosis, genetic counselling, and medical management have improved. These advances in medical practice have occurred while definitive therapies based on an improved knowledge of disease pathogenesis are awaited. A wide range of therapeutic options have been tested in animal models, and some are being tested in clinical trials. Various therapeutic targets are being investigated, from personalised medicines targeting specific mutations and drugs targeting cellular pathways to gene-based and cell-based therapies.
Collapse
Affiliation(s)
- Kate Bushby
- Institute of Human Genetics, Newcastle University, Newcastle upon Tyne, UK.
| | | | | | | |
Collapse
|
46
|
The distribution and characterization of skeletal muscle lesions in dysferlin-deficient SJL and A/J mice. ACTA ACUST UNITED AC 2009; 62:509-17. [PMID: 19615872 DOI: 10.1016/j.etp.2009.06.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2009] [Accepted: 06/27/2009] [Indexed: 11/23/2022]
Abstract
The pathogenesis of limb-girdle muscular dystrophy type 2B (LGMD2B) dysferlinopathy remains to be investigated. The distribution and characterization of skeletal muscle lesions were examined in two different LGMD2B mouse models, SJL and A/J mice (at 10 and 35 weeks old), in association with the endoplasmic reticulum (ER) stress. SJL mice showed an earlier age of onset and a faster progression of skeletal muscle lesions as compared with those of A/J mice; the sensitivity difference to muscular dystrophic lesions between SJL and A/J mice was observed in the lumbar muscles (particularly, lumbar longissimus and sublumbar muscles); the lesions seen mainly in SJL mice at 35 weeks old consisted of degeneration, necrosis, fatty infiltration, variation in muscle fiber size and atrophy in muscle fibers. Enzyme-histochemically, the fast-twitch muscle fiber was predominant for the degenerative changes seen in the rectus femoris and lateral longissimus muscles of SJL mice. Immunohistochemically, the main reactive cell type observed in and around degenerative and/or necrotic muscle fibers was macrophages, demonstrable with an anti-F4/80 antibody. Because the analyses of spliced XBP1 mRNA, a marker of ER stress, did not show the increased expression, it was considered that ER stress did not affect the progression of skeletal muscle lesions in SJL mice with the advanced stage of dysferlinopathy.
Collapse
|
47
|
Hochmeister S, Bittner RE, Höger H, Lassmann H, Bradl M. The susceptibility to experimental autoimmune encephalomyelitis is not related to dysferlin-deficiency. Autoimmunity 2009; 42:235-41. [PMID: 19301206 DOI: 10.1080/08916930802716542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Recent observations suggested that dysferlin might play a role in the development of autoimmune central nervous system (CNS) inflammation. To address this issue, we studied the induction and effector phase of experimental autoimmune encephalomyelitis in C57BL/10 mice producing intact or functionally deficient dysferlin. We found that both types of mice showed identical T-cell and antibody responses against the immunogen, and developed CNS inflammation with identical clinical courses, frequencies, lesion distributions, sizes and compositions. These findings suggest that the presence or absence of dysferlin does not have any consequences for the triggering or effector phase of autoimmune CNS inflammation.
Collapse
Affiliation(s)
- Sonja Hochmeister
- Department of Neuroimmunology, Center for Brain Reseach, Medical University Vienna, Austria
| | | | | | | | | |
Collapse
|
48
|
De la Torre C, Illa I, Faulkner G, Soria L, Robles-Cedeño R, Dominguez-Perles R, De Luna N, Gallardo E. Proteomics identification of differentially expressed proteins in the muscle of dysferlin myopathy patients. Proteomics Clin Appl 2009; 3:486-97. [PMID: 21136973 DOI: 10.1002/prca.200800087] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2008] [Indexed: 01/16/2023]
Abstract
The muscular dystrophies are a large and heterogeneous group of neuromuscular disorders that can be classified according to the mode of inheritance, the clinical phenotype and the molecular defect. To better understand the pathological mechanisms of dysferlin myopathy we compared the protein-expression pattern in the muscle biopsies of six patients with this disease with six patients with limb girdle muscular dystrophy 2A, five with facioscapulohumeral dystrophy and six normal control subjects. To investigate differences in the expression levels of skeletal muscle proteins we used 2-DE and MS. Western blot or immunohistochemistry confirmed relevant results. The study showed specific increase expression of proteins involved in fast-to-slow fiber type conversion (ankyrin repeat protein 2), type I predominance (phosphorylated forms of slow troponin T), sarcomere stabilization (actinin-associated LIM protein), protein ubiquitination (TRIM 72) and skeletal muscle differentiation (Rho-GDP-dissociation inhibitor ly-GDI) in dysferlin myopathy. As anticipated, we also found differential expression of proteins common to all the muscular dystrophies studied. This comparative proteomic analysis suggests that in dysferlin myopathy (i) the type I fiber predominance is an active process of fiber type conversion rather than a selective loss of type II fibers and (ii) the dysregulation of proteins involved in muscle differentiation further confirms the role of dysferlin in this process.
Collapse
Affiliation(s)
- Carolina De la Torre
- Laboratory of Experimental Neurology, Institut de Recerca HSCSP, Universitat Autònoma de Barcelona, Barcelona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Cheong JK, Gunaratnam L, Zang ZJ, Yang CM, Sun X, Nasr SL, Sim KG, Peh BK, Rashid SBA, Bonventre JV, Salto-Tellez M, Hsu SI. TRIP-Br2 promotes oncogenesis in nude mice and is frequently overexpressed in multiple human tumors. J Transl Med 2009; 7:8. [PMID: 19152710 PMCID: PMC2671481 DOI: 10.1186/1479-5876-7-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2008] [Accepted: 01/20/2009] [Indexed: 01/09/2023] Open
Abstract
Background Members of the TRIP-Br/SERTAD family of mammalian transcriptional coregulators have recently been implicated in E2F-mediated cell cycle progression and tumorigenesis. We, herein, focus on the detailed functional characterization of the least understood member of the TRIP-Br/SERTAD protein family, TRIP-Br2 (SERTAD2). Methods Oncogenic potential of TRIP-Br2 was demonstrated by (1) inoculation of NIH3T3 fibroblasts, which were engineered to stably overexpress ectopic TRIP-Br2, into athymic nude mice for tumor induction and (2) comprehensive immunohistochemical high-throughput screening of TRIP-Br2 protein expression in multiple human tumor cell lines and human tumor tissue microarrays (TMAs). Clinicopathologic analysis was conducted to assess the potential of TRIP-Br2 as a novel prognostic marker of human cancer. RNA interference of TRIP-Br2 expression in HCT-116 colorectal carcinoma cells was performed to determine the potential of TRIP-Br2 as a novel chemotherapeutic drug target. Results Overexpression of TRIP-Br2 is sufficient to transform murine fibroblasts and promotes tumorigenesis in nude mice. The transformed phenotype is characterized by deregulation of the E2F/DP-transcriptional pathway through upregulation of the key E2F-responsive genes CYCLIN E, CYCLIN A2, CDC6 and DHFR. TRIP-Br2 is frequently overexpressed in both cancer cell lines and multiple human tumors. Clinicopathologic correlation indicates that overexpression of TRIP-Br2 in hepatocellular carcinoma is associated with a worse clinical outcome by Kaplan-Meier survival analysis. Small interfering RNA-mediated (siRNA) knockdown of TRIP-Br2 was sufficient to inhibit cell-autonomous growth of HCT-116 cells in vitro. Conclusion This study identifies TRIP-Br2 as a bona-fide protooncogene and supports the potential for TRIP-Br2 as a novel prognostic marker and a chemotherapeutic drug target in human cancer.
Collapse
Affiliation(s)
- Jit Kong Cheong
- Renal Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Blaauw B, Mammucari C, Toniolo L, Agatea L, Abraham R, Sandri M, Reggiani C, Schiaffino S. Akt activation prevents the force drop induced by eccentric contractions in dystrophin-deficient skeletal muscle. Hum Mol Genet 2008; 17:3686-96. [DOI: 10.1093/hmg/ddn264] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
|