The immune system in Duchenne muscular dystrophy: Friend or foe

Duchenne muscular dystrophy (DMD) is a genetic disease caused by mutations in the X-linked dystrophin gene, resulting in reduced or absent protein production, subsequently leading to the structural instability of the dystroglycan complex (DGC), muscle degeneration, and early death in males. Thus, current treatments have been targeting the genetic defect either by bypassing the mutation through exon skipping or replacing the defective gene through gene therapy and stem cell approaches. However, what has been an underappreciated mediator of muscle pathology and, ultimately, of muscle degeneration and fibrotic replacement, is the prominent inflammatory response. Of potentially critical importance, however, is the fact that the elements mediating the inflammatory response also play an essential role in tissue repair. In this opinion piece, we highlight the detrimental and supportive immune parameters that occur as a consequence of the genetic disorder and discuss how changes to immunity can potentially ameliorate the disease intensity and be employed in conjunction with efforts to correct the genetic disorder.

[1]  A. Sadeghi,et al.  Redefinition of dystrophin isoform distribution in mouse tissue by RT-PCR implies role in nonmuscle manifestations of duchenne muscular dystrophy. , 1998, Molecular genetics and metabolism.

[2]  E. Hoffman,et al.  Osteopontin promotes fibrosis in dystrophic mouse muscle by modulating immune cell subsets and intramuscular TGF-beta. , 2009, The Journal of clinical investigation.

[3]  I. Nonaka,et al.  Inflammatory response in facioscapulohumeral muscular dystrophy (FSHD): Immunocytochemical and genetic analyses , 1995, Muscle & nerve. Supplement.

[4]  J. Tidball,et al.  A nitric oxide synthase transgene ameliorates muscular dystrophy in mdx mice , 2001, The Journal of cell biology.

[5]  Bernard Dan,et al.  Pathophysiology of duchenne muscular dystrophy: current hypotheses. , 2007, Pediatric neurology.

[6]  J. Kern,et al.  The T‐cell receptor repertoire of regulatory T cells , 2008, Immunology.

[7]  J. Sullivan,et al.  Interleukin 35: A Key Mediator of Suppression and the Propagation of Infectious Tolerance , 2013, Front. Immunol..

[8]  T. Partridge,et al.  T-Cell-Dependent Fibrosis in the mdx Dystrophic Mouse , 2000, Laboratory Investigation.

[9]  J. Mendell,et al.  Anti-dystrophin T cell responses in Duchenne muscular dystrophy: prevalence and a glucocorticoid treatment effect. , 2013, Human gene therapy.

[10]  C. Reggiani,et al.  Inflammation in muscular dystrophy and the beneficial effects of non‐steroidal anti‐inflammatory drugs , 2012, Muscle & nerve.

[11]  R. Mantegazza,et al.  Fibrosis and inflammation are greater in muscles of beta-sarcoglycan-null mouse than mdx mouse , 2014, Cell and Tissue Research.

[12]  Mark M Davis,et al.  Virus-specific CD4(+) memory-phenotype T cells are abundant in unexposed adults. , 2013, Immunity.

[13]  K. L. Gardner,et al.  Interplay of IKK/NF-kappaB signaling in macrophages and myofibers promotes muscle degeneration in Duchenne muscular dystrophy. , 2007, The Journal of clinical investigation.

[14]  J. Tidball,et al.  Helper (CD4(+)) and cytotoxic (CD8(+)) T cells promote the pathology of dystrophin-deficient muscle. , 2001, Clinical immunology.

[15]  Raffaele Iorio,et al.  CD8+ T Cells in Facioscapulohumeral Muscular Dystrophy Patients with Inflammatory Features at Muscle MRI , 2011, Journal of Clinical Immunology.

[16]  A. Manzur,et al.  Glucocorticoid corticosteroids for Duchenne muscular dystrophy. , 2004, The Cochrane database of systematic reviews.

[17]  S. Batonnet-Pichon,et al.  Srf-dependent paracrine signals produced by myofibers control satellite cell-mediated skeletal muscle hypertrophy. , 2012, Cell metabolism.

[18]  M. Yano,et al.  Interleukin 6 receptor antibody inhibits muscle atrophy and modulates proteolytic systems in interleukin 6 transgenic mice. , 1996, The Journal of clinical investigation.

[19]  H. Sweeney,et al.  Long-term administration of the TNF blocking drug Remicade (cV1q) to mdx mice reduces skeletal and cardiac muscle fibrosis, but negatively impacts cardiac function , 2014, Neuromuscular Disorders.

[20]  A. Pristerá,et al.  Tumor Necrosis Factor‐α Inhibition of Skeletal Muscle Regeneration Is Mediated by a Caspase‐Dependent Stem Cell Response , 2008, Stem cells.

[21]  S. Goerdt,et al.  Differences in angiogenic potential of classically vs alternatively activated macrophages. , 1997, Immunobiology.

[22]  Phoebe Lin,et al.  The future of uveitis treatment. , 2014, Ophthalmology.

[23]  J. Myśliwska,et al.  First-in-man clinical results of the treatment of patients with graft versus host disease with human ex vivo expanded CD4+CD25+CD127- T regulatory cells. , 2009, Clinical immunology.

[24]  S. Gordon Alternative activation of macrophages , 2003, Nature Reviews Immunology.

[25]  J. Buckner,et al.  IL-2 therapy in type 1 diabetes: "Trials" and tribulations. , 2013, Clinical immunology.

[26]  J. Tidball,et al.  Do immune cells promote the pathology of dystrophin-deficient myopathies? , 2001, Neuromuscular Disorders.

[27]  J. Bluestone,et al.  Control of peripheral T‐cell tolerance and autoimmunity via the CTLA‐4 and PD‐1 pathways , 2008, Immunological reviews.

[28]  J. Tidball,et al.  IFN-γ Promotes Muscle Damage in the mdx Mouse Model of Duchenne Muscular Dystrophy by Suppressing M2 Macrophage Activation and Inhibiting Muscle Cell Proliferation , 2011, The Journal of Immunology.

[29]  J. Bluestone,et al.  Regulatory T cells suppress muscle inflammation and injury in muscular dystrophy , 2014, Science Translational Medicine.

[30]  K. Flanigan,et al.  A Comparative Study of N-glycolylneuraminic Acid (Neu5Gc) and Cytotoxic T Cell (CT) Carbohydrate Expression in Normal and Dystrophin-Deficient Dog and Human Skeletal Muscle , 2014, PloS one.

[31]  Yi-Wen Chen,et al.  IL-6 signaling blockade increases inflammation but does not affect muscle function in the mdx mouse , 2012, BMC Musculoskeletal Disorders.

[32]  A. Musarò,et al.  Muscle-specific expression of insulin-like growth factor I counters muscle decline in mdx mice , 2002, The Journal of cell biology.

[33]  J. Bluestone,et al.  T Regulatory Cells in Autoimmune Diabetes: Past Challenges, Future Prospects , 2008, Journal of Clinical Immunology.

[34]  M. Grounds,et al.  Reduced muscle necrosis and long-term benefits in dystrophic mdx mice after cV1q (blockade of TNF) treatment , 2008, Neuromuscular Disorders.

[35]  H. Hartung,et al.  Fingolimod is a potential novel therapy for multiple sclerosis , 2010, Nature Reviews Neurology.

[36]  W. Młynarski,et al.  Administration of CD4+CD25highCD127− Regulatory T Cells Preserves β-Cell Function in Type 1 Diabetes in Children , 2012, Diabetes Care.

[37]  J. Ritz,et al.  Interleukin-2 and regulatory T cells in graft-versus-host disease. , 2011, The New England journal of medicine.

[38]  Abraham P. Fong,et al.  DUX4 activates germline genes, retroelements, and immune mediators: implications for facioscapulohumeral dystrophy. , 2012, Developmental cell.

[39]  A. Engel,et al.  Monoclonal antibody analysis of mononuclear cells in myopathies. I: Quantitation of subsets according to diagnosis and sites of accumulation and demonstration and counts of muscle fibers invaded by T cells , 1984, Annals of neurology.

[40]  J. Tidball,et al.  Shared signaling systems in myeloid cell-mediated muscle regeneration , 2014, Development.

[41]  M. Grounds,et al.  Reduced necrosis of dystrophic muscle by depletion of host neutrophils, or blocking TNFα function with Etanercept in mdx mice , 2006, Neuromuscular Disorders.

[42]  C. Miao,et al.  In vivo expansion of regulatory T cells with IL-2/IL-2 mAb complexes prevents anti-factor VIII immune responses in hemophilia A mice treated with factor VIII plasmid-mediated gene therapy. , 2011, Molecular therapy : the journal of the American Society of Gene Therapy.

[43]  J. Tidball,et al.  Interleukin-10 reduces the pathology of mdx muscular dystrophy by deactivating M1 macrophages and modulating macrophage phenotype. , 2011, Human molecular genetics.

[44]  B. Paepe,et al.  Cytokines and Chemokines as Regulators of Skeletal Muscle Inflammation: Presenting the Case of Duchenne Muscular Dystrophy , 2013 .

[45]  B. De Paepe,et al.  Cytokines and Chemokines as Regulators of Skeletal Muscle Inflammation: Presenting the Case of Duchenne Muscular Dystrophy , 2013, Mediators of inflammation.

[46]  L. Tseng-Ong,et al.  Eosinophilia of dystrophin-deficient muscle is promoted by perforin-mediated cytotoxicity by T cell effectors. , 2000, The American journal of pathology.

[47]  M. Monden,et al.  Muscle undergoes atrophy in association with increase of lysosomal cathepsin activity in interleukin-6 transgenic mouse. , 1995, Biochemical and biophysical research communications.

[48]  H. Blau,et al.  Specific T cell receptor gene rearrangements at the site of muscle degeneration in Duchenne muscular dystrophy. , 1994, Journal of immunology.

[49]  T. Gotoh,et al.  Shifts in macrophage phenotypes and macrophage competition for arginine metabolism affect the severity of muscle pathology in muscular dystrophy. , 2008, Human molecular genetics.

[50]  N. F. Kamel,et al.  The effects of glucocorticoid therapy on the inflammatory and Dendritic cells in muscular dystrophies , 2006, International journal of experimental pathology.

[51]  James J. Lee,et al.  Major basic protein-1 promotes fibrosis of dystrophic muscle and attenuates the cellular immune response in muscular dystrophy. , 2008, Human molecular genetics.

[52]  J. Mendell,et al.  Dystrophin immunity in Duchenne's muscular dystrophy. , 2010, The New England journal of medicine.

[53]  Y. Ohira,et al.  Anti-interleukin-6 receptor antibody (MR16-1) promotes muscle regeneration via modulation of gene expressions in infiltrated macrophages. , 2014, Biochimica et biophysica acta.

[54]  Mark M. Davis,et al.  Deconstructing the Peptide-MHC Specificity of T Cell Recognition , 2014, Cell.

[55]  G. Hansson,et al.  Treg-mediated suppression of atherosclerosis requires MYD88 signaling in DCs. , 2013, The Journal of clinical investigation.

[56]  L. Partridge,et al.  Signaling by IL-6 promotes alternative activation of macrophages to limit endotoxemia and obesity-associated resistance to insulin , 2014, Nature Immunology.

[57]  J. Bluestone,et al.  Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo , 2009, Nature Immunology.

[58]  James M. Wilson Autoimmunity, recessive diseases, and gene replacement therapy. , 2010, Molecular therapy : the journal of the American Society of Gene Therapy.

[59]  Andrew C. Chan,et al.  Therapeutic antibodies for autoimmunity and inflammation , 2010, Nature Reviews Immunology.

[60]  A. Manzur,et al.  Glucocorticoid corticosteroids for Duchenne muscular dystrophy. , 2008, The Cochrane database of systematic reviews.

[61]  Qizhi Tang,et al.  The Foxp3+ regulatory T cell: a jack of all trades, master of regulation , 2008, Nature Immunology.

[62]  L. Steinman Four easy pieces : Interconnections between tissue injury, intermediary metabolism, autoimmunity, and chronic degeneration , 2006 .

[63]  M. Krumbholz,et al.  B cells and antibodies in multiple sclerosis pathogenesis and therapy , 2012, Nature Reviews Neurology.

[64]  James J. Lee,et al.  Prednisolone decreases cellular adhesion molecules required for inflammatory cell infiltration in dystrophin-deficient skeletal muscle , 2004, Neuromuscular Disorders.

[65]  L. Skov,et al.  Targeting of interleukin-17 in the treatment of psoriasis , 2014, Clinical, cosmetic and investigational dermatology.

[66]  C. Benoist,et al.  A Special Population of Regulatory T Cells Potentiates Muscle Repair , 2013, Cell.

[67]  Yoshiya Tanaka,et al.  IL-6 targeting compared to TNF targeting in rheumatoid arthritis: studies of olokizumab, sarilumab and sirukumab , 2014, Annals of the rheumatic diseases.

[68]  Yi-Ping Li,et al.  TNF-α regulates myogenesis and muscle regeneration by activating p38 MAPK , 2007 .

[69]  B. Pedersen,et al.  Interleukin‐6 myokine signaling in skeletal muscle: a double‐edged sword? , 2013, The FEBS journal.

[70]  Akinori Nakamura,et al.  Evaluation of dystrophic dog pathology by fat‐suppressed T2‐weighted imaging , 2009, Muscle & nerve.

[71]  K. Moore,et al.  X chromosome-linked muscular dystrophy (mdx) in the mouse. , 1984, Proceedings of the National Academy of Sciences of the United States of America.

[72]  G. D’Antona,et al.  T and B lymphocyte depletion has a marked effect on the fibrosis of dystrophic skeletal muscles in the scid/mdx mouse , 2007, The Journal of pathology.

[73]  J. Tidball,et al.  Regulatory interactions between muscle and the immune system during muscle regeneration. , 2010, American journal of physiology. Regulatory, integrative and comparative physiology.

[74]  P. Muñoz-Cánoves,et al.  Interleukin-6 is an essential regulator of satellite cell-mediated skeletal muscle hypertrophy. , 2008, Cell metabolism.