1
|
Lauletta A, Allenbach Y, Béhin A, Evangelista T, Léonard-Louis S, Garibaldi M, Benveniste O. High prevalence of facioscapulohumeral muscular dystrophy (FSHD) and inflammatory myopathies association: Is there an interplay? J Neurol Sci 2025; 470:123400. [PMID: 39855012 DOI: 10.1016/j.jns.2025.123400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 12/19/2024] [Accepted: 01/18/2025] [Indexed: 01/27/2025]
Abstract
INTRODUCTION Certain types of muscular dystrophy (MD), notably facioscapulohumeral muscular dystrophy (FSHD), exhibit muscle fiber necrosis with regeneration and a nonspecific inflammatory process. Although rare, the coexistence of MDs and autoimmune myositis has been observed. We hypothesized that, in some circumstances, FSHD may predispose individuals to myositis through muscle damage-induced autoantigen overexpression, contributing to an autoimmune response. METHODS We conducted a retrospective analysis of patient data from neuromuscular disease centers in France and Italy between September 2012 and May 2024. Clinical, immunological, and myopathological features of 1750 myositis patients were comprehensively reviewed. RESULTS Five patients were identified with both FSHD and IIM. Two patients were first diagnosed with FSHD and later developed IIM, while two others initially had IIM followed by an FSHD diagnosis. The fifth patient received simultaneous diagnoses of both conditions. The prevalence of FSHD in the IIM cohort was 1/350, and the prevalence of IIM in the FSHD cohort was 1/40 (p < 0.0001). DISCUSSION Our study showed a high prevalence of FSHD and IIM association compared to the general population, with underlying mechanisms that remain unclear. This association might be more frequent than previously reported, indicating a need for increased clinical awareness. Understanding the interplay between FSHD and autoimmune myositis could reveal insights into the immunopathological processes of these diseases and improve diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Antonio Lauletta
- Neuromuscular Disease Centre, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy.
| | - Yves Allenbach
- Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Inserm U974, Department of Internal Medicine and Clinical Immunology, Pitié-Salpêtrière University Hospital, Paris, France
| | - Anthony Béhin
- APHP, Centre de Référence des Maladies Neuromusculaires Nord/Est/Ile de France, Institut de Myologie, Hôpital Pitié-Salpêtrière, Paris, France
| | - Teresinha Evangelista
- Unité de Morphologie Neuromusculaire, Institut de Myologie, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - Sarah Léonard-Louis
- Service de Neuromyologie, GH Pitié-Salpêtrière, University Hospital, 75013 Paris, France
| | - Matteo Garibaldi
- Neuromuscular Disease Centre, Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Sapienza University of Rome, Sant'Andrea Hospital, Rome, Italy
| | - Olivier Benveniste
- Sorbonne Université, Assistance Publique - Hôpitaux de Paris, Inserm U974, Department of Internal Medicine and Clinical Immunology, Pitié-Salpêtrière University Hospital, Paris, France.
| |
Collapse
|
2
|
Pacilio S, Lombardi S, Costa R, Paris F, Petrocelli G, Marrazzo P, Cenacchi G, Alviano F. Role of Perinatal Stem Cell Secretome as Potential Therapy for Muscular Dystrophies. Biomedicines 2025; 13:458. [PMID: 40002871 PMCID: PMC11852414 DOI: 10.3390/biomedicines13020458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/02/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammation mechanisms play a critical role in muscle homeostasis, and in Muscular Dystrophies (MDs), the myofiber damage triggers chronic inflammation which significantly controls the disease progression. Immunomodulatory strategies able to target inflammatory pathways and mitigate the immune-mediated damage in MDs may provide new therapeutic options. Owing to its capacity of influencing the immune response and enhancing tissue repair, stem cells' secretome has been proposed as an adjunct or standalone treatment for MDs. In this review study, we discuss the challenging points related to the inflammation condition characterizing MD pathology and provide a concise summary of the literature supporting the potential of perinatal stem cells in targeting and modulating the MD inflammation.
Collapse
Affiliation(s)
- Serafina Pacilio
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Sara Lombardi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Roberta Costa
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Francesca Paris
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Giovannamaria Petrocelli
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy;
| | - Pasquale Marrazzo
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, 61029 Urbino, Italy
| | - Giovanna Cenacchi
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| | - Francesco Alviano
- Department of Biomedical and Neuromotor Sciences, Alma Mater Studiorum University of Bologna, 40126 Bologna, Italy; (S.P.); (S.L.); (R.C.); (F.P.); (G.C.); (F.A.)
| |
Collapse
|
3
|
Hermes TDA, Fratini P, Nascimento BG, Ferreira LL, Petri G, Fonseca FLA, Carvalho AADS, Feder D. Trilobatin contributes to the improvement of myopathy in a mouse model of Duchenne muscular dystrophy. Int J Exp Pathol 2024; 105:75-85. [PMID: 38477495 PMCID: PMC10951423 DOI: 10.1111/iep.12502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 02/06/2024] [Accepted: 02/13/2024] [Indexed: 03/14/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) occurs due to genetic mutations that lead to a deficiency in dystrophin production and consequent progressive degeneration of skeletal muscle fibres, through oxidative stress and an exacerbated inflammatory process. The flavonoid trilobatin (TLB) demonstrates antioxidant and anti-inflammatory potential. Its high safety profile and effective action make it a potent therapy for the process of dystrophic muscle myonecrosis. Thus, we sought to investigate the action of TLB on damage in a DMD model, the mdx mouse. Eight-week-old male animals were treated with 160 mg/kg/day of trilobatin for 8 weeks. Control animals were treated with saline. Following treatment, muscle strength, serum creatine kinase (CK) levels, histopathology (necrotic myofibres, regenerated fibres/central nuclei, Feret's diameter and inflammatory area) and the levels of catalase and NF-κB (western blotting) of the quadriceps (QUA), diaphragm (DIA) and tibialis anterior (TA) muscles were measured. TLB was able to significantly increase muscle strength and reduce serum CK levels in dystrophic animals. The QUA of mdx mice showed a reduction in catalase and the number of fibres with a centralized nucleus after treatment with TLB. In the DIA of dystrophic animals, TLB reduced the necrotic myofibres, inflammatory area and NF-κB and increased the number of regenerated fibres and the total fibre diameter. In TA, TLB increased the number of regenerated fibres and reduced catalase levels in these animals. It is concluded that in the mdx experimental model, treatment with TLB was beneficial in the treatment of DMD.
Collapse
Affiliation(s)
- Túlio de Almeida Hermes
- Department of Anatomy, ICBFederal University of Alfenas (UNIFAL‐MG)AlfenasMinas GeraisBrazil
| | - Paula Fratini
- Department of PharmacologyCentro Universitário FMABC (FMABC)Santo AndréSao PauloBrazil
| | | | - Laís Leite Ferreira
- Department of Anatomy, ICBFederal University of Alfenas (UNIFAL‐MG)AlfenasMinas GeraisBrazil
| | - Giuliana Petri
- Department of PharmacologyCentro Universitário FMABC (FMABC)Santo AndréSao PauloBrazil
| | | | | | - David Feder
- Department of PharmacologyCentro Universitário FMABC (FMABC)Santo AndréSao PauloBrazil
| |
Collapse
|
4
|
Mozzetta C, Sartorelli V, Steinkuhler C, Puri PL. HDAC inhibitors as pharmacological treatment for Duchenne muscular dystrophy: a discovery journey from bench to patients. Trends Mol Med 2024; 30:278-294. [PMID: 38408879 PMCID: PMC11095976 DOI: 10.1016/j.molmed.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 01/22/2024] [Accepted: 01/24/2024] [Indexed: 02/28/2024]
Abstract
Earlier evidence that targeting the balance between histone acetyltransferases (HATs) and deacetylases (HDACs), through exposure to HDAC inhibitors (HDACis), could enhance skeletal myogenesis, prompted interest in using HDACis to promote muscle regeneration. Further identification of constitutive HDAC activation in dystrophin-deficient muscles, caused by dysregulated nitric oxide (NO) signaling, provided the rationale for HDACi-based therapeutic interventions for Duchenne muscular dystrophy (DMD). In this review, we describe the molecular, preclinical, and clinical evidence supporting the efficacy of HDACis in countering disease progression by targeting pathogenic networks of gene expression in multiple muscle-resident cell types of patients with DMD. Given that givinostat is paving the way for HDACi-based interventions in DMD, next-generation HDACis with optimized therapeutic profiles and efficacy could be also explored for synergistic combinations with other therapeutic strategies.
Collapse
Affiliation(s)
- Chiara Mozzetta
- Institute of Molecular Biology and Pathology (IBPM), National Research Council (CNR) of Italy, Rome, Italy
| | - Vittorio Sartorelli
- Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases (NIAMS), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Pier Lorenzo Puri
- Development, Aging, and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.
| |
Collapse
|
5
|
The Immune System in Duchenne Muscular Dystrophy Pathogenesis. Biomedicines 2021; 9:biomedicines9101447. [PMID: 34680564 PMCID: PMC8533196 DOI: 10.3390/biomedicines9101447] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 12/24/2022] Open
Abstract
Growing evidence demonstrates the crosstalk between the immune system and the skeletal muscle in inflammatory muscle diseases and dystrophic conditions such as Duchenne Muscular Dystrophy (DMD), as well as during normal muscle regeneration. The rising of inflammation and the consequent activation of the immune system are hallmarks of DMD: several efforts identified the immune cells that invade skeletal muscle as CD4+ and CD8+ T cells, Tregs, macrophages, eosinophils and natural killer T cells. The severity of muscle injury and inflammation dictates the impairment of muscle regeneration and the successive replacement of myofibers with connective and adipose tissue. Since immune system activation was traditionally considered as a consequence of muscular wasting, we recently demonstrated a defect in central tolerance caused by thymus alteration and the presence of autoreactive T-lymphocytes in DMD. Although the study of innate and adaptive immune responses and their complex relationship in DMD attracted the interest of many researchers in the last years, the results are so far barely exhaustive and sometimes contradictory. In this review, we describe the most recent improvements in the knowledge of immune system involvement in DMD pathogenesis, leading to new opportunities from a clinical point-of-view.
Collapse
|
6
|
A Systematic Review on the Role of SIRT1 in Duchenne Muscular Dystrophy. Cells 2021; 10:cells10061380. [PMID: 34205021 PMCID: PMC8229470 DOI: 10.3390/cells10061380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 05/26/2021] [Accepted: 05/31/2021] [Indexed: 12/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a muscular disease characterized by progressive muscle degeneration. Life expectancy is between 30 and 50 years, and death is correlated with cardiac or respiratory complications. Currently, there is no cure, so there is a great interest in new pharmacological targets. Sirtuin1 (SIRT1) seems to be a potential target for DMD. In muscle tissue, SIRT1 exerts anti-inflammatory and antioxidant effects. The aim of this study is to summarize all the findings of in vivo and in vitro literature studies about the potential role of SIRT1 in DMD. A systematic literature search was performed according to PRISMA guidelines. Twenty-three articles satisfied the eligibility criteria. It emerged that SIRT1 inhibition led to muscle fragility, while conversely its activation improved muscle function. Additionally, resveratrol, a SIRT1 activator, has brought beneficial effects to the skeletal, cardiac and respiratory muscles by exerting anti-inflammatory activity that leads to reduced myofiber wasting.
Collapse
|
7
|
Xing T, Luo D, Zhao X, Xu X, Li J, Zhang L, Gao F. Enhanced cytokine expression and upregulation of inflammatory signaling pathways in broiler chickens affected by wooden breast myopathy. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:279-286. [PMID: 32623748 DOI: 10.1002/jsfa.10641] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 06/21/2020] [Accepted: 07/05/2020] [Indexed: 06/11/2023]
Abstract
BACKGROUND Wooden breast (WB) myopathy in broiler chickens is a growing challenge for the poultry industry. Previous multi-omic data have implied that the pathogenesis of WB is associated with the activation of immune system and inflammatory response. However, the intricate mechanisms are not fully understood. This study was therefore conducted to systematically investigate the morphology, expression of cytokines as well as the underlying signaling pathways regulating the inflammatory response in pectoralis major (PM) muscle of WB myopathic broilers. RESULTS wHistopathological changes, increased plasma creatine kinase and lactate dehydrogenase activities, elevated myeloperoxidase activity and overproduction of nitric oxide in muscle indicated the enhancement of muscle damage and inflammation in WB broilers. The messenger RNA (mRNA) expressions of inflammatory cytokines were dysregulated in PM muscle and contents of interleukin (IL)-1β, IL-8 and tumor necrosis factor-α were increased in serum of WB myopathic broilers, indicating this myopathy was associated with immune disorder and systemic inflammation response. Additionally, toll-like receptor (TLR) levels were upregulated, the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway was activated and the mRNA expression levels of downstream inflammatory mediators were enhanced in PM muscle of WB myopathy affected birds. CONCLUSION The results indicated an immune disorder and a systemic inflammation response in WB myopathic broilers, which might be related to a synergetic effect of TLRs and NF-κB pathway. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Tong Xing
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, P. R. China
| | - Dan Luo
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, P. R. China
| | - Xue Zhao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, P. R. China
| | - Xinglian Xu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, P. R. China
| | - Jiaolong Li
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, P. R. China
| | - Lin Zhang
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, P. R. China
| | - Feng Gao
- College of Animal Science and Technology, Key Laboratory of Animal Origin Food Production and Safety Guarantee of Jiangsu Province, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Joint International Research Laboratory of Animal Health and Food Safety, National Experimental Teaching Demonstration Center of Animal Science, Nanjing Agricultural University, Nanjing, P. R. China
| |
Collapse
|
8
|
Lira FS, Conrado de Freitas M, Gerosa-Neto J, Cholewa JM, Rossi FE. Comparison Between Full-Body vs. Split-Body Resistance Exercise on the Brain-Derived Neurotrophic Factor and Immunometabolic Response. J Strength Cond Res 2020; 34:3094-3102. [PMID: 33105359 DOI: 10.1519/jsc.0000000000002653] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Lira, FS, Conrado de Freitas, M, Gerosa-Neto, J, Cholewa, JM, and Rossi, FE. Comparison between full-body vs. split-body resistance exercise on the brain-derived neurotrophic factor immunometabolic response. J Strength Cond Res 34(11): 3094-3102, 2020-Intense aerobic exercise seems to increase serum concentrations of brain-derived neurotrophic factor (BDNF) in conjunction with increasing lactate; however, less is known about the BDNF response to differing resistance exercise protocols. We hypothesized that full-body (FB) resistance exercise will elicit a greater increase in serum BDNF and lactate compared with split-body resistance exercise. Twelve recreationally resistance-trained men (age = 25.3 ± 5.9 years) performed 3 randomized trials of 18 sets of exercise: upper-body (UB), lower-body (LB), and FB conditions. Serum BDNF levels were assessed at rest, immediately Post-exercise, Post-1 hour, and Post-2 hours during recovery. Lactate concentration was evaluated at rest, after 9 sets, Post-exercise, Post-5, Post-10, and Post-30 minutes during recovery. In addition, interleukin (IL-6 and IL-10) and the IL-6/IL-10 ratio were calculated. Lactate concentration and total volume were greater in the FB condition compared with LB and UB (p < 0.05). For BDNF, effect sizes were largest in the LB (1.4), followed by the FB (0.75), and moderate to UB (0.33), although no significant differences were observed between conditions. There was a statistically significant relationship between lactate and BDNF only for LB condition (rho = 0.72; p = 0.013). There were a greater IL-10 Post-1 hour for FB condition compared with UB and LB (p < 0.001), and lower IL-6/IL-10 ratio in FB compared with UB (p < 0.001). Lower body induced a great BDNF response, and FB resistance exercise elicited a greater increase of serum cytokines than UB in trained men. We speculate that the volume of work performed by larger muscles has a larger influence on BDNF than overall volume.
Collapse
Affiliation(s)
- Fabio S Lira
- Department of Physical Education, Exercise and Immunometabolism Research Group, São Paulo State University (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Marcelo Conrado de Freitas
- Department of Physical Education, Skeletal Muscle Assessment Laboratory, School of Technology and Sciences, São Paulo State University, Presidente Prudente, São Paulo, Brazil
| | - Jose Gerosa-Neto
- Department of Physical Education, Exercise and Immunometabolism Research Group, São Paulo State University (UNESP), Presidente Prudente, São Paulo, Brazil
| | - Jason M Cholewa
- Department of Kinesiology, Coastal Carolina University, Conway, South Carolina; and
| | - Fabrício E Rossi
- Department of Physical Education, Immunometabolism of Skeletal Muscle and Exercise Research Group, Federal University of Piauí (UFPI), Teresina, Brazil
| |
Collapse
|
9
|
Suntar I, Sureda A, Belwal T, Sanches Silva A, Vacca RA, Tewari D, Sobarzo-Sánchez E, Nabavi SF, Shirooie S, Dehpour AR, Xu S, Yousefi B, Majidinia M, Daglia M, D'Antona G, Nabavi SM. Natural products, PGC-1 α , and Duchenne muscular dystrophy. Acta Pharm Sin B 2020; 10:734-745. [PMID: 32528825 PMCID: PMC7276681 DOI: 10.1016/j.apsb.2020.01.001] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 10/14/2019] [Accepted: 12/06/2019] [Indexed: 02/08/2023] Open
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a transcriptional coactivator that binds to a diverse range of transcription factors. PPARγ coactivator 1 (PGC-1) coactivators possess an extensive range of biological effects in different tissues, and play a key part in the regulation of the oxidative metabolism, consequently modulating the production of reactive oxygen species, autophagy, and mitochondrial biogenesis. Owing to these findings, a large body of studies, aiming to establish the role of PGC-1 in the neuromuscular system, has shown that PGC-1 could be a promising target for therapies targeting neuromuscular diseases. Among these, some evidence has shown that various signaling pathways linked to PGC-1α are deregulated in muscular dystrophy, leading to a reduced capacity for mitochondrial oxidative phosphorylation and increased reactive oxygen species (ROS) production. In the light of these results, any intervention aimed at activating PGC-1 could contribute towards ameliorating the progression of muscular dystrophies. PGC-1α is influenced by different patho-physiological/pharmacological stimuli. Natural products have been reported to display modulatory effects on PPARγ activation with fewer side effects in comparison to synthetic drugs. Taken together, this review summarizes the current knowledge on Duchenne muscular dystrophy, focusing on the potential effects of natural compounds, acting as regulators of PGC-1α.
Collapse
Key Words
- AAV, adeno-associated virus
- AMP, adenosine monophosphate
- AMPK, 5′ adenosine monophosphate-activated protein kinase
- ASO, antisense oligonucleotides
- ATF2, activating transcription factor 2
- ATP, adenosine triphosphate
- BMD, Becker muscular dystrophy
- COPD, chronic obstructive pulmonary disease
- CREB, cyclic AMP response element-binding protein
- CnA, calcineurin a
- DAGC, dystrophin-associated glycoprotein complex
- DGC, dystrophin–glycoprotein complex
- DMD, Duchenne muscular dystrophy
- DRP1, dynamin-related protein 1
- DS, Down syndrome
- ECM, extracellular matrix
- EGCG, epigallocatechin-3-gallate
- ERRα, estrogen-related receptor alpha
- FDA, U. S. Food and Drug Administration
- FGF, fibroblast growth factor
- FOXO1, forkhead box class-O1
- GABP, GA-binding protein
- GPX, glutathione peroxidase
- GSK3b, glycogen synthase kinase 3b
- HCT, hydrochlorothiazide
- HDAC, histone deacetylase
- HIF-1α, hypoxia-inducible factors
- IL, interleukin
- LDH, lactate dehydrogenase
- MCP-1, monocyte chemoattractant protein-1
- MD, muscular dystrophy
- MEF2, myocyte enhancer factor 2
- MSCs, mesenchymal stem cells
- Mitochondrial oxidative phosphorylation
- Muscular dystrophy
- MyoD, myogenic differentiation
- NADPH, nicotinamide adenine dinucleotide phosphate
- NF-κB, nuclear factor kappa-light-chain-enhancer of activated B cells
- NMJ, neuromuscular junctions
- NO, nitric oxide
- NOS, NO synthase
- Natural product
- PDGF, platelet derived growth factor
- PGC-1, peroxisome proliferator-activated receptor γ coactivator 1
- PPARγ activation
- PPARγ, peroxisome proliferator-activated receptor γ
- Peroxisome proliferator-activated receptor γ coactivator 1α
- ROS, reactive oxygen species
- Reactive oxygen species
- SIRT1, silent mating type information regulation 2 homolog 1
- SOD, superoxide dismutase
- SPP1, secreted phosphoprotein 1
- TNF-α, tumor necrosis factor-α
- UCP, uncoupling protein
- VEGF, vascular endothelial growth factor
- cGMP, cyclic guanosine monophosphate
- iPSCs, induced pluripotent stem cells
- p38 MAPK, p38 mitogen-activated protein kinase
Collapse
|
10
|
Saclier M, Lapi M, Bonfanti C, Rossi G, Antonini S, Messina G. The Transcription Factor Nfix Requires RhoA-ROCK1 Dependent Phagocytosis to Mediate Macrophage Skewing during Skeletal Muscle Regeneration. Cells 2020; 9:E708. [PMID: 32183151 PMCID: PMC7140652 DOI: 10.3390/cells9030708] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 03/11/2020] [Accepted: 03/12/2020] [Indexed: 01/13/2023] Open
Abstract
Macrophages (MPs) are immune cells which are crucial for tissue repair. In skeletal muscle regeneration, pro-inflammatory cells first infiltrate to promote myogenic cell proliferation, then they switch into an anti-inflammatory phenotype to sustain myogenic cells differentiation and myofiber formation. This phenotypical switch is induced by dead cell phagocytosis. We previously demonstrated that the transcription factor Nfix, a member of the nuclear factor I (Nfi) family, plays a pivotal role during muscle development, regeneration and in the progression of muscular dystrophies. Here, we show that Nfix is mainly expressed by anti-inflammatory macrophages. Upon acute injury, mice deleted for Nfix in myeloid line displayed a significant defect in the process of muscle regeneration. Indeed, Nfix is involved in the macrophage phenotypical switch and macrophages lacking Nfix failed to adopt an anti-inflammatory phenotype and interact with myogenic cells. Moreover, we demonstrated that phagocytosis induced by the inhibition of the RhoA-ROCK1 pathway leads to Nfix expression and, consequently, to acquisition of the anti-inflammatory phenotype. Our study identified Nfix as a link between RhoA-ROCK1-dependent phagocytosis and the MP phenotypical switch, thus establishing a new role for Nfix in macrophage biology for the resolution of inflammation and tissue repair.
Collapse
Affiliation(s)
| | | | | | | | | | - Graziella Messina
- Department of Biosciences, University of Milan, via Celoria 26, 20133 Milan, Italy; (M.S.); (M.L.); (C.B.); (G.R.); (S.A.)
| |
Collapse
|
11
|
Murata I, Imanari M, Komiya M, Kobayashi J, Inoue Y, Kanamoto I. Icing treatment in rats with crush syndrome can improve survival through reduction of potassium concentration and mitochondrial function disorder effect. Exp Ther Med 2019; 19:777-785. [PMID: 31853328 DOI: 10.3892/etm.2019.8230] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 05/08/2019] [Indexed: 12/12/2022] Open
Abstract
Crush syndrome (CS), a serious medical condition, which is characterized by damage to myocytes due to pressure and is associated with high mortality, even when patients receive fluid therapy. Icing therapy over the affected muscle has been reported to be effective in improving mitochondrial dysfunction and inflammation. These effects are thought to be secondary to improvements in the leakage of potassium and myoglobin from the damaged myocytes in the early stages of disease. However, their effects on the various symptoms of CS are unclear. It was hypothesized that treatment with icing will inhibit the influence of potassium by vasoconstriction, exert anti-inflammatory effects in the affected myocytes and improve mitochondrial function The CS model constructed by subjecting anesthetized rats to bilateral hindlimb compression with a rubber tourniquet for 5 h. The rats were then randomly divided into six groups: i) Sham; ii) CS without treatment (CS); iii) and iv) icing for 30 or 180 min over the entire hindlimb on CS rats (CI-30 and -180), respectively; and v) and vi) local icing for 30 or 180 min over the affected area on CS rats (CLI-30 and -180), respectively. Under continuous monitoring and recording of arterial blood pressures, blood and tissue samples were collected for biochemical analyses at designated time points prior to and following reperfusion. The survival rate, vital signs, and blood gas parameters in the CS group were lethal compared with the sham group. These were also improved in the CI-30 and CLI-30 groups compared with the CS group; however, they worsened in the CI-180 and CLI-180 groups due to hypothermia. The CI-30 and CLI-30 groups demonstrated tendencies of improvements compared with the CS group. Systemic inflammation and mitochondria dysfunction had improved in these groups compared with the CS group. We suggest icing therapy to temporarily prolong the viability after crush injury. Its effectiveness can be improved by combining it with other infusion therapies.
Collapse
Affiliation(s)
- Isamu Murata
- Laboratory of Drug Safety Management, Faculty of Pharmacy and Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| | - Mayuki Imanari
- Laboratory of Drug Safety Management, Faculty of Pharmacy and Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| | - Marise Komiya
- Laboratory of Drug Safety Management, Faculty of Pharmacy and Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| | - Jun Kobayashi
- Division of Pathophysiology, Department of Clinical Dietetics and Human Nutrition, Faculty of Pharmacy and Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| | - Yutaka Inoue
- Laboratory of Drug Safety Management, Faculty of Pharmacy and Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| | - Ikuo Kanamoto
- Laboratory of Drug Safety Management, Faculty of Pharmacy and Pharmaceutical Science, Josai University, Sakado, Saitama 350-0295, Japan
| |
Collapse
|
12
|
Differential diagnosis of idiopathic inflammatory myopathies in adults - the first step when approaching a patient with muscle weakness. Reumatologia 2018; 56:307-315. [PMID: 30505013 PMCID: PMC6263305 DOI: 10.5114/reum.2018.79502] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 09/25/2018] [Indexed: 11/17/2022] Open
Abstract
Despite its misleading adjective, the most commonly used diagnostic criteria of idiopathic inflammatory myopathies (IIM) are applicable only after all other non-autoimmune muscle diseases have been excluded. It makes differential diagnosis the first step when approaching a patient with muscle weakness. This article is designed to list the most common conditions from which to differentiate in rheumatological care. In fact, many patients with the diseases described here have been initially misdiagnosed with IIM. For the purpose of this article, only the most commonly found and important conditions according to the authors are listed with the essence of information; other autoimmune muscle diseases, such as sarcoidosis and eosinophilic myositis, are not portrayed. The attached bibliography may serve as a source, when further exploration of a specific subject is needed.
Collapse
|
13
|
Mashinchian O, Pisconti A, Le Moal E, Bentzinger CF. The Muscle Stem Cell Niche in Health and Disease. Curr Top Dev Biol 2017; 126:23-65. [PMID: 29305000 DOI: 10.1016/bs.ctdb.2017.08.003] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The regulation of stem cells that maintain and regenerate postnatal tissues depends on extrinsic signals originating from their microenvironment, commonly referred to as the stem cell niche. Complex higher-order regulatory interrelationships with the tissue and factors in the systemic circulation are integrated and propagated to the stem cells through the niche. The stem cell niche in skeletal muscle tissue is both a paradigm for a structurally and functionally relatively static niche that maintains stem cell quiescence during tissue homeostasis, and a highly dynamic regenerative niche that is subject to extensive structural remodeling and a flux of different support cell populations. Conditions ranging from aging to chronically degenerative skeletal muscle diseases affect the composition of the niche and thereby impair the regenerative potential of muscle stem cells. A holistic and integrative understanding of the extrinsic mechanisms regulating muscle stem cells in health and disease in a broad systemic context will be imperative for the identification of regulatory hubs in the niche interactome that can be targeted to maintain, restore, or enhance the regenerative capacity of muscle tissue. Here, we review the microenvironmental regulation of muscle stem cells, summarize how niche dysfunction can contribute to disease, and discuss emerging therapeutic implications.
Collapse
Affiliation(s)
- Omid Mashinchian
- Nestlé Institute of Health Sciences, Lausanne, Switzerland; École Polytechnique Fédérale de Lausanne, Doctoral Program in Biotechnology and Bioengineering, Lausanne, Switzerland
| | - Addolorata Pisconti
- Institute of Integrative Biology, University of Liverpool, Liverpool, United Kingdom
| | - Emmeran Le Moal
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada
| | - C Florian Bentzinger
- Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, QC, Canada.
| |
Collapse
|
14
|
Significant response to immune therapies in a case of subacute necrotizing myopathy and FKRP mutations. Neuromuscul Disord 2015; 25:865-8. [PMID: 26363967 DOI: 10.1016/j.nmd.2015.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 08/16/2015] [Accepted: 08/19/2015] [Indexed: 10/23/2022]
Abstract
Necrotizing myopathies can be encountered in various conditions as acquired myopathies (toxic or autoimmune) or muscular dystrophies. We report a twenty-year-old Caucasian woman who presented with clinical findings suggestive of an inflammatory myopathy: subacute onset of lower limb muscle weakness, myalgia, weight loss and absence of family history. The serum creatine kinase level was elevated at 4738 IU/L (normal range, 25-175 IU/L). Muscle biopsy was consistent with necrotizing myopathy. The patient showed significant clinical improvement following corticosteroid, azathioprine and intravenous immunoglobulin treatments. Biological tests revealed no specific autoantibodies associated with necrotizing autoimmune myopathies. Immunohistochemical staining for sarcolemmal proteins in muscle biopsy samples finally led to a diagnosis of limb-girdle muscular dystrophy 2I (fukutin-related protein gene mutations). The response to immune therapies suggested a possible inflammatory component associated with the muscular dystrophy and highlighted the potential benefit of corticosteroid treatment in patients with LGMD2I and subacute onset.
Collapse
|
15
|
Artlett CM, Sassi-Gaha S, Ramos RC, Miller FW, Rider LG. Chimeric cells of maternal origin do not appear to be pathogenic in the juvenile idiopathic inflammatory myopathies or muscular dystrophy. Arthritis Res Ther 2015; 17:238. [PMID: 26338728 PMCID: PMC4558637 DOI: 10.1186/s13075-015-0732-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Accepted: 07/28/2015] [Indexed: 01/08/2023] Open
Abstract
Introduction Microchimeric cells have been studied for over a decade, with conflicting reports on their presence and role in autoimmune and other inflammatory diseases. To determine whether microchimeric cells were pathogenic or mediating tissue repair in inflammatory myopathies, we phenotyped and quantified microchimeric cells in juvenile idiopathic inflammatory myopathies (JIIM), muscular dystrophy (MD), and noninflammatory control muscle tissues. Method Fluorescence immunophenotyping for infiltrating cells with sequential fluorescence in situ hybridization was performed on muscle biopsies from ten patients with JIIM, nine with MD and ten controls. Results Microchimeric cells were significantly increased in MD muscle (0.079 ± 0.024 microchimeric cells/mm2 tissue) compared to controls (0.019 ± 0.007 cells/mm2 tissue, p = 0.01), but not elevated in JIIM muscle (0.043 ± 0.015 cells/mm2). Significantly more CD4+ and CD8+ microchimeric cells were in the muscle of patients with MD compared with controls (mean 0.053 ± 0.020/mm2 versus 0 ± 0/mm2p = 0.003 and 0.043 ± 0.023/mm2 versus 0 ± 0/mm2p = 0.025, respectively). No differences in microchimeric cells between JIIM, MD, and noninflammatory controls were found for CD3+, Class II+, CD25+, CD45RA+, and CD123+ phenotypes, and no microchimeric cells were detected in CD20, CD83, or CD45RO populations. The locations of microchimeric cells were similar in all three conditions, with MD muscle having more microchimeric cells in perimysial regions than controls, and JIIM having fewer microchimeric muscle nuclei than MD. Microchimeric inflammatory cells were found, in most cases, at significantly lower proportions than autologous cells of the same phenotype. Conclusions Microchimeric cells are not specific to autoimmune disease, and may not be important in muscle inflammation or tissue repair in JIIM.
Collapse
Affiliation(s)
- Carol M Artlett
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA, 19129, USA.
| | - Sihem Sassi-Gaha
- Department of Microbiology and Immunology, Drexel University College of Medicine, 2900 Queen Lane, Philadelphia, PA, 19129, USA.
| | - Ronald C Ramos
- Department of Medicine, Thomas Jefferson University, 1020 Walnut Street, Philadelphia, PA, 19107, USA. .,Present address: Agis Global, Business Development Executive, 1266 East Main Street, Stamford, CT, 06902, USA.
| | - Frederick W Miller
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Environmental Autoimmunity Group, Program of Clinical Research, 9000 Rockville Pike, Bethesda, MD, 20892, USA.
| | - Lisa G Rider
- National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Environmental Autoimmunity Group, Program of Clinical Research, 9000 Rockville Pike, Bethesda, MD, 20892, USA.
| |
Collapse
|
16
|
Sun CC, Li SJ, Yang CL, Xue RL, Xi YY, Wang L, Zhao QL, Li DJ. Sulforaphane Attenuates Muscle Inflammation in Dystrophin-deficient mdx Mice via NF-E2-related Factor 2 (Nrf2)-mediated Inhibition of NF-κB Signaling Pathway. J Biol Chem 2015; 290:17784-17795. [PMID: 26013831 PMCID: PMC4505027 DOI: 10.1074/jbc.m115.655019] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Revised: 05/11/2015] [Indexed: 12/22/2022] Open
Abstract
Inflammation is widely distributed in patients with Duchenne muscular dystrophy and ultimately leads to progressive deterioration of muscle function with chronic muscle damage, oxidative stress, and reduced oxidative capacity. NF-E2-related factor 2 (Nrf2) plays a critical role in defending against inflammation in different tissues via activation of phase II enzyme heme oxygenase-1 and inhibition of the NF-κB signaling pathway. However, the role of Nrf2 in the inflammation of dystrophic muscle remains unknown. To determine whether Nrf2 may counteract inflammation in dystrophic muscle, we treated 4-week-old male mdx mice with the Nrf2 activator sulforaphane (SFN) by gavage (2 mg/kg of body weight/day) for 4 weeks. The experimental results demonstrated that SFN treatment increased the expression of muscle phase II enzyme heme oxygenase-1 in an Nrf2-dependent manner. Inflammation in mice was reduced by SFN treatment as indicated by decreased infiltration of immune cells and expression of the inflammatory cytokine CD45 and proinflammatory cytokines tumor necrosis factor-α, interleukin-1β, and interleukin-6 in the skeletal muscles of mdx mice. In addition, SFN treatment also decreased the expression of NF-κB(p65) and phosphorylated IκB kinase-α as well as increased inhibitor of κB-α expression in mdx mice in an Nrf2-dependent manner. Collectively, these results show that SFN-induced Nrf2 can alleviate muscle inflammation in mdx mice by inhibiting the NF-κB signaling pathway.
Collapse
Affiliation(s)
- Cheng-Cao Sun
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, China
| | - Shu-Jun Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, China
| | - Cui-Li Yang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, China
| | - Rui-Lin Xue
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, China
| | - Yong-Yong Xi
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, China
| | - Liang Wang
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, China
| | - Qian-Long Zhao
- Department of Occupational and Environmental Health, School of Public Health, Lanzhou University, 730000 Lanzhou, China
| | - De-Jia Li
- Department of Occupational and Environmental Health, School of Public Health, Wuhan University, 430071 Wuhan, China.
| |
Collapse
|
17
|
FOXP3+ T Cells Recruited to Sites of Sterile Skeletal Muscle Injury Regulate the Fate of Satellite Cells and Guide Effective Tissue Regeneration. PLoS One 2015; 10:e0128094. [PMID: 26039259 PMCID: PMC4454513 DOI: 10.1371/journal.pone.0128094] [Citation(s) in RCA: 137] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2015] [Accepted: 04/23/2015] [Indexed: 01/07/2023] Open
Abstract
Muscle injury induces a classical inflammatory response in which cells of the innate immune system rapidly invade the tissue. Macrophages are prominently involved in this response and required for proper healing, as they are known to be important for clearing cellular debris and supporting satellite cell differentiation. Here, we sought to assess the role of the adaptive immune system in muscle regeneration after acute damage. We show that T lymphocytes are transiently recruited into the muscle after damage and appear to exert a pro-myogenic effect on muscle repair. We observed a decrease in the cross-sectional area of regenerating myofibers after injury in Rag2-/- γ-chain-/- mice, as compared to WT controls, suggesting that T cell recruitment promotes muscle regeneration. Skeletal muscle infiltrating T lymphocytes were enriched in CD4+CD25+FOXP3+ cells. Direct exposure of muscle satellite cells to in vitro induced Treg cells effectively enhanced their expansion, and concurrently inhibited their myogenic differentiation. In vivo, the recruitment of Tregs to acutely injured muscle was limited to the time period of satellite expansion, with possibly important implications for situations in which inflammatory conditions persist, such as muscular dystrophies and inflammatory myopathies. We conclude that the adaptive immune system, in particular T regulatory cells, is critically involved in effective skeletal muscle regeneration. Thus, in addition to their well-established role as regulators of the immune/inflammatory response, T regulatory cells also regulate the activity of skeletal muscle precursor cells, and are instrumental for the proper regeneration of this tissue.
Collapse
|
18
|
Pelosi L, Coggi A, Forcina L, Musarò A. MicroRNAs modulated by local mIGF-1 expression in mdx dystrophic mice. Front Aging Neurosci 2015; 7:69. [PMID: 25999854 PMCID: PMC4419723 DOI: 10.3389/fnagi.2015.00069] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Accepted: 04/20/2015] [Indexed: 01/22/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a X-linked genetic disease in which the absence of dystrophin leads to progressive lethal skeletal muscle degeneration. It has been demonstrated that among genes which are important for proper muscle development and function, micro-RNAs (miRNAs) play a crucial role. Moreover, altered levels of miRNAs were found in several muscular disorders, including DMD. A specific group of miRNAs, whose expression depends on dystrophin levels and whose deregulation explains several DMD pathogenetic traits, has been identified. Here, we addressed whether the anabolic activity of mIGF-1 on dystrophic muscle is associated with modulation of microRNAs expression. We demonstrated that some microRNAs are strictly linked to the dystrophin expression and are not modulated by mIGF-1 expression. In contrast, local expression of mIGF-1 promotes the modulation of other microRNAs, such as miR-206 and miR-24, along with the modulation of muscle specific genes, which are associated with maturation of regenerating fibers and with the stabilization of the differentiated muscle phenotype. These data suggest that mIGF-1, modifying the expression of some of the active players of muscle homeostasis, is able, even in absence of dystrophin expression, to activate circuitries that confer robustness to dystrophic muscle.
Collapse
Affiliation(s)
- Laura Pelosi
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM, Sapienza University of Rome Rome, Italy
| | - Angela Coggi
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM, Sapienza University of Rome Rome, Italy
| | - Laura Forcina
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM, Sapienza University of Rome Rome, Italy
| | - Antonio Musarò
- Institute Pasteur Cenci-Bolognetti, DAHFMO-Unit of Histology and Medical Embryology, IIM, Sapienza University of Rome Rome, Italy ; Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia Rome, Italy
| |
Collapse
|
19
|
Villalta SA, Rosenthal W, Martinez L, Kaur A, Sparwasser T, Tidball JG, Margeta M, Spencer MJ, Bluestone JA. Regulatory T cells suppress muscle inflammation and injury in muscular dystrophy. Sci Transl Med 2014; 6:258ra142. [PMID: 25320234 PMCID: PMC4889432 DOI: 10.1126/scitranslmed.3009925] [Citation(s) in RCA: 193] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
We examined the hypothesis that regulatory T cells (Tregs) modulate muscle injury and inflammation in the mdx mouse model of Duchenne muscular dystrophy (DMD). Although Tregs were largely absent in the muscle of wild-type mice and normal human muscle, they were present in necrotic lesions, displayed an activated phenotype, and showed increased expression of interleukin-10 (IL-10) in dystrophic muscle from mdx mice. Depletion of Tregs exacerbated muscle injury and the severity of muscle inflammation, which was characterized by an enhanced interferon-γ (IFN-γ) response and activation of M1 macrophages. To test the therapeutic value of targeting Tregs in muscular dystrophy, we treated mdx mice with IL-2/anti-IL-2 complexes and found that Tregs and IL-10 concentrations were increased in muscle, resulting in reduced expression of cyclooxygenase-2 and decreased myofiber injury. These findings suggest that Tregs modulate the progression of muscular dystrophy by suppressing type 1 inflammation in muscle associated with muscle fiber injury, and highlight the potential of Treg-modulating agents as therapeutics for DMD.
Collapse
Affiliation(s)
- S Armando Villalta
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA.
| | - Wendy Rosenthal
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Leonel Martinez
- Department of Neurology and Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Amanjot Kaur
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Tim Sparwasser
- Institute for Infection Immunology, Twincore, Hannover 30625, Germany
| | - James G Tidball
- Molecular, Cellular, and Integrative Physiology Program, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Marta Margeta
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Melissa J Spencer
- Department of Neurology and Center for Duchenne Muscular Dystrophy, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jeffrey A Bluestone
- Diabetes Center, University of California, San Francisco, San Francisco, CA 94143, USA. Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA. Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
20
|
Rouillon J, Zocevic A, Leger T, Garcia C, Camadro JM, Udd B, Wong B, Servais L, Voit T, Svinartchouk F. Proteomics profiling of urine reveals specific titin fragments as biomarkers of Duchenne muscular dystrophy. Neuromuscul Disord 2014; 24:563-73. [DOI: 10.1016/j.nmd.2014.03.012] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 03/12/2014] [Accepted: 03/28/2014] [Indexed: 11/30/2022]
|
21
|
Bryant ND, Li K, Does MD, Barnes S, Gochberg DF, Yankeelov TE, Park JH, Damon BM. Multi-parametric MRI characterization of inflammation in murine skeletal muscle. NMR IN BIOMEDICINE 2014; 27:716-25. [PMID: 24777935 PMCID: PMC4134016 DOI: 10.1002/nbm.3113] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2013] [Revised: 02/10/2014] [Accepted: 03/13/2014] [Indexed: 05/15/2023]
Abstract
Myopathies often display a common set of complex pathologies that include muscle weakness, inflammation, compromised membrane integrity, fat deposition, and fibrosis. Multi-parametric, quantitative, non-invasive imaging approaches may be able to resolve these individual pathological components. The goal of this study was to use multi-parametric MRI to investigate inflammation as an isolated pathological feature. Proton relaxation, diffusion tensor imaging (DTI), quantitative magnetization transfer (qMT-MRI), and dynamic contrast enhanced (DCE-MRI) parameters were calculated from data acquired in a single imaging session conducted 6-8 hours following the injection of λ-carrageenan, a local inflammatory agent. T2 increased in the inflamed muscle and transitioned to bi-exponential behavior. In diffusion measurements, all three eigenvalues and the apparent diffusion coefficient increased, but λ3 had the largest relative change. Analysis of the qMT data revealed that the T1 of the free pool and the observed T1 both increased in the inflamed tissue, while the ratio of exchanging spins in the solid pool to those in the free water pool (the pool size ratio) significantly decreased. DCE-MRI data also supported observations of an increase in extracellular volume. These findings enriched the understanding of the relation between multiple quantitative MRI parameters and an isolated inflammatory pathology, and may potentially be employed for other single or complex myopathy models.
Collapse
Affiliation(s)
- Nathan D Bryant
- Vanderbilt University Institute of Imaging Science, Vanderbilt University, Nashville, TN, USA; Department of Radiology and Radiological Sciences, Vanderbilt University, Nashville, TN, USA
| | | | | | | | | | | | | | | |
Collapse
|
22
|
van de Vyver M, Myburgh KH. Variable inflammation and intramuscular STAT3 phosphorylation and myeloperoxidase levels after downhill running. Scand J Med Sci Sports 2014; 24:e360-71. [PMID: 24383415 DOI: 10.1111/sms.12164] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2013] [Indexed: 12/30/2022]
Abstract
Individual responses in creatine kinase (CK) release after eccentric exercise are divergent. This study aimed to identify whether this could be related to selected humoral or intramuscular inflammatory factors. Twenty-three subjects were divided into non-exercising (n = 5) and downhill run (DHR; n = 18) groups (12 × 5 min, 10% decline at 15 km/h). Blood samples were analyzed for white blood cell differential count, CK, myoglobin, tumor necrosis factor-α, granulocyte colony-stimulating factor, interleukin (IL)-1β, IL-6, and IL-10. Muscle biopsies were analyzed for signal transducer and activator of transcription-3 (STAT3), IκBα, and myeloperoxidase (MPO). DHR participants clustered as early (DHR1) recovery, biphasic response (DHR2), or classic delayed exaggerated CK response (DHR3), with a delayed CK peak (4784 ± 1496 U/L) on day 4. For DHR1 and DHR2, CK peaked on day 1 (DHR1: 1198 ± 837 U/L) or on day 1 and day 4 (DHR2: 1583 ± 448 U/L; 1878 ± 427 U/L), respectively. Immediately post-DHR, IL-6 increased in DHR2 and DHR3 whereas IL-10 increased in all DHR groups. STAT3 signaling increased for DHR1 and DHR2 at 4 h, but MPO at day 2 only in DHR2. Objective cluster analysis uncovered a group of subjects with a characteristic biphasic CK release after DHR. The second elevation was related to their early cytokine response. The results provide evidence that early responses following eccentric exercise are indicative of later variation.
Collapse
Affiliation(s)
- M van de Vyver
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | | |
Collapse
|
23
|
De Paepe B, De Bleecker JL. Cytokines and chemokines as regulators of skeletal muscle inflammation: presenting the case of Duchenne muscular dystrophy. Mediators Inflamm 2013; 2013:540370. [PMID: 24302815 PMCID: PMC3835490 DOI: 10.1155/2013/540370] [Citation(s) in RCA: 90] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2013] [Accepted: 09/09/2013] [Indexed: 01/09/2023] Open
Abstract
Duchenne muscular dystrophy is a severe inherited muscle disease that affects 1 in 3500 boys worldwide. Infiltration of skeletal muscle by inflammatory cells is an important facet of disease pathophysiology and is strongly associated with disease severity in the individual patient. In the chronic inflammation that characterizes Duchenne muscle, cytokines and chemokines are considered essential activators and recruiters of inflammatory cells. In addition, they provide potential beneficiary effects on muscle fiber damage control and tissue regeneration. In this review, current knowledge of cytokine and chemokine expression in Duchenne muscular dystrophy and its relevant animal disease models is listed, and implications for future therapeutic avenues are discussed.
Collapse
Affiliation(s)
- Boel De Paepe
- Laboratory for Myopathology, Department of Neurology and Neuromuscular Reference Center, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| | - Jan L. De Bleecker
- Laboratory for Myopathology, Department of Neurology and Neuromuscular Reference Center, Ghent University Hospital, De Pintelaan 185, 9000 Ghent, Belgium
| |
Collapse
|
24
|
Mancuso M, Orsucci D, Ienco EC, Ricci G, Ali G, Servadio A, Fontanini G, Filosto M, Vielmi V, Rocchi A, Petrozzi L, LoGerfo A, Siciliano G. An “inflammatory” mitochondrial myopathy. A case report. Neuromuscul Disord 2013; 23:907-10. [DOI: 10.1016/j.nmd.2013.07.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Revised: 06/07/2013] [Accepted: 07/31/2013] [Indexed: 01/01/2023]
|
25
|
Hindi SM, Sato S, Choi Y, Kumar A. Distinct roles of TRAF6 at early and late stages of muscle pathology in the mdx model of Duchenne muscular dystrophy. Hum Mol Genet 2013; 23:1492-505. [PMID: 24163132 DOI: 10.1093/hmg/ddt536] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal genetic disorder caused by loss of functional dystrophin protein. Accumulating evidence suggests that the deficiency of dystrophin leads to aberrant activation of many signaling pathways which contribute to disease progression. However, the proximal signaling events leading to the activation of various pathological cascades in dystrophic muscle remain less clear. TNF receptor-associated factor 6 (TRAF6) is an adaptor protein which acts as a signaling intermediate for several receptor-mediated signaling events leading to the context-dependent activation of a number of signaling pathways. TRAF6 is also an E3 ubiquitin ligase and an important regulator of autophagy. However, the role of TRAF6 in pathogenesis of DMD remains unknown. Here, we demonstrate that the levels and activity of TRAF6 are increased in skeletal muscle of mdx (a mouse model of DMD) mice. Targeted deletion of TRAF6 improves muscle strength and reduces fiber necrosis, infiltration of macrophages and the activation of proinflammatory transcription factor nuclear factor-kappa B (NF-κB) in 7-week-old mdx mice. Ablation of TRAF6 also increases satellite cells proliferation and myofiber regeneration in young mdx mice. Intriguingly, ablation of TRAF6 exacerbates muscle injury and increases fibrosis in 9-month-old mdx mice. TRAF6 inhibition reduces the markers of autophagy and Akt signaling in dystrophic muscle of mdx mice. Collectively, our study suggests that while the inhibition of TRAF6 improves muscle structure and function in young mdx mice, its continued inhibition causes more severe myopathy at later stages of disease progression potentially through repressing autophagy.
Collapse
Affiliation(s)
- Sajedah M Hindi
- Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA and
| | | | | | | |
Collapse
|
26
|
Gene expression profiling identifies molecular pathways associated with collagen VI deficiency and provides novel therapeutic targets. PLoS One 2013; 8:e77430. [PMID: 24223098 PMCID: PMC3819505 DOI: 10.1371/journal.pone.0077430] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 09/02/2013] [Indexed: 12/25/2022] Open
Abstract
Ullrich congenital muscular dystrophy (UCMD), caused by collagen VI deficiency, is a common congenital muscular dystrophy. At present, the role of collagen VI in muscle and the mechanism of disease are not fully understood. To address this we have applied microarrays to analyse the transcriptome of UCMD muscle and compare it to healthy muscle and other muscular dystrophies. We identified 389 genes which are differentially regulated in UCMD relative to controls. In addition, there were 718 genes differentially expressed between UCMD and dystrophin deficient muscle. In contrast, only 29 genes were altered relative to other congenital muscular dystrophies. Changes in gene expression were confirmed by real-time PCR. The set of regulated genes was analysed by Gene Ontology, KEGG pathways and Ingenuity Pathway analysis to reveal the molecular functions and gene networks associated with collagen VI defects. The most significantly regulated pathways were those involved in muscle regeneration, extracellular matrix remodelling and inflammation. We characterised the immune response in UCMD biopsies as being mainly mediated via M2 macrophages and the complement pathway indicating that anti-inflammatory treatment may be beneficial to UCMD as for other dystrophies. We studied the immunolocalisation of ECM components and found that biglycan, a collagen VI interacting proteoglycan, was reduced in the basal lamina of UCMD patients. We propose that biglycan reduction is secondary to collagen VI loss and that it may be contributing towards UCMD pathophysiology. Consequently, strategies aimed at over-expressing biglycan and restore the link between the muscle cell surface and the extracellular matrix should be considered.
Collapse
|
27
|
Percival JM, Whitehead NP, Adams ME, Adamo CM, Beavo JA, Froehner SC. Sildenafil reduces respiratory muscle weakness and fibrosis in the mdx mouse model of Duchenne muscular dystrophy. J Pathol 2012; 228:77-87. [PMID: 22653783 DOI: 10.1002/path.4054] [Citation(s) in RCA: 120] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Revised: 05/16/2010] [Accepted: 05/21/2010] [Indexed: 11/07/2022]
Abstract
Duchenne muscular dystrophy (DMD) is the most common form of muscular dystrophy caused by mutations in the dystrophin gene. Loss of dystrophin initiates a progressive decline in skeletal muscle integrity and contractile capacity which weakens respiratory muscles including the diaphragm, culminating in respiratory failure, the leading cause of morbidity and mortality in DMD patients. At present, corticosteroid treatment is the primary pharmacological intervention in DMD, but has limited efficacy and adverse side effects. Thus, there is an urgent need for new safe, cost-effective, and rapidly implementable treatments that slow disease progression. One promising new approach is the amplification of nitric oxide-cyclic guanosine monophosphate (NO-cGMP) signalling pathways with phosphodiesterase 5 (PDE5) inhibitors. PDE5 inhibitors serve to amplify NO signalling that is attenuated in many neuromuscular diseases including DMD. We report here that a 14-week treatment of the mdx mouse model of DMD with the PDE5 inhibitor sildenafil (Viagra(®), Revatio(®)) significantly reduced mdx diaphragm muscle weakness without impacting fatigue resistance. In addition to enhancing respiratory muscle contractility, sildenafil also promoted normal extracellular matrix organization. PDE5 inhibition slowed the establishment of mdx diaphragm fibrosis and reduced matrix metalloproteinase-13 (MMP-13) expression. Sildenafil also normalized the expression of the pro-fibrotic (and pro-inflammatory) cytokine tumour necrosis factor α (TNFα). Sildenafil-treated mdx diaphragms accumulated significantly less Evans Blue tracer dye than untreated controls, which is also indicative of improved diaphragm muscle health. We conclude that sildenafil-mediated PDE5 inhibition significantly reduces diaphragm respiratory muscle dysfunction and pathology in the mdx mouse model of Duchenne muscular dystrophy. This study provides new insights into the therapeutic utility of targeting defects in NO-cGMP signalling with PDE5 inhibitors in dystrophin-deficient muscle.
Collapse
Affiliation(s)
- Justin M Percival
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195, USA.
| | | | | | | | | | | |
Collapse
|
28
|
Gordon BS, Delgado Díaz DC, Kostek MC. Resveratrol decreases inflammation and increases utrophin gene expression in the mdx mouse model of Duchenne muscular dystrophy. Clin Nutr 2012; 32:104-11. [PMID: 22795790 DOI: 10.1016/j.clnu.2012.06.003] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 05/23/2012] [Accepted: 06/04/2012] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS Duchenne muscular dystrophy (DMD) is a lethal genetic disease with no cure. Reducing inflammation or increasing utrophin expression can alleviate DMD pathology. Resveratrol can reduce inflammation and activate the utrophin promoter. The aims of this study were to identify an active dose of resveratrol in mdx mice and examine if this dose decreased inflammation and increased utrophin expression. METHODS 5-week old mdx mice were given 0, 10, 100, or 500 mg/kg of resveratrol everyday for 10 days. Sirt1 was measured by qRT-PCR and used to determine the most active dose. Muscle inflammation was measured by H&E staining, CD45 and F4/80 immunohistochemistry. IL-6, TNFα, PGC-1α, and utrophin gene expression were measured by qRT-PCR. Utrophin, Sirt1, and PGC-1α protein were quantified by western blot. RESULTS The 100 mg/kg dose of resveratrol, the most active dose, increased Sirt1 mRNA 60 ± 10% (p < 0.01), reduced immune cell infiltration 21 ± 6% (H&E) and 42 ± 8% (CD45 immunohistochemistry (p < 0.05)), reduced macrophage infiltration 48 ± 10% (F4/80 immunohistochemistry (p < 0.05)), and increased IL-6, PGC-1α, and utrophin mRNA 247 ± 77%, 27 ± 17%, and 43 ± 23% respectively (p ≤ 0.05). Utrophin, Sirt1, and PGC-1α protein expression did not change. CONCLUSIONS Resveratrol may be a therapy for DMD by reducing inflammation.
Collapse
Affiliation(s)
- Bradley S Gordon
- Department of Exercise Science, University of South Carolina, Columbia, SC 29208, USA
| | | | | |
Collapse
|
29
|
Kim JH, Lawler JM. Amplification of proinflammatory phenotype, damage, and weakness by oxidative stress in the diaphragm muscle of mdx mice. Free Radic Biol Med 2012; 52:1597-606. [PMID: 22330042 DOI: 10.1016/j.freeradbiomed.2012.01.015] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2011] [Revised: 01/10/2012] [Accepted: 01/20/2012] [Indexed: 12/27/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a common and devastating type of childhood-onset muscular dystrophy, attributed to an X-linked defect in the gene that encodes dystrophin. Myopathy with DMD is most pronounced in the diaphragm muscle and fast-twitch limb muscles and is dependent upon susceptibility to damage, inflammatory cell infiltration, and proinflammatory signaling (nuclear factor-κB; NF-κB). Although recent papers have reawakened the notion that oxidative stress links inflammatory signaling with pathology in DMD in limb muscle, the importance of redox mechanisms had been clouded by inconsistent results from indirect scavenger approaches, including in the diaphragm muscle. Therefore, we used a novel catalytic mimetic of superoxide dismutase and catalase (EUK-134) as a direct scavenger of oxidative stress in myopathy in the diaphragm of the mdx mouse model. EUK-134 reduced 4-hydroxynonenal and total hydroperoxides, markers of oxidative stress in the mdx diaphragm. EUK-134 also attenuated positive staining of macrophages and T-cells as well as activation of NF-κB and p65 protein abundance. Moreover, EUK-134 ameliorated markers of muscle damage including internalized nuclei, variability of cross-sectional area, and type IIc fibers. Finally, impairment of contractile force was partially rescued by EUK-134 in the diaphragm of mdx mice. We conclude that oxidative stress amplifies DMD pathology in the diaphragm muscle.
Collapse
Affiliation(s)
- Jong-Hee Kim
- Redox Biology and Cell Signaling Laboratory, Department of Health & Kinesiology, Texas A&M University, College Station, TX 77843-4243, USA
| | | |
Collapse
|
30
|
TNF-Alpha in the Locomotor System beyond Joints: High Degree of Involvement in Myositis in a Rabbit Model. Int J Rheumatol 2012; 2012:637452. [PMID: 22505941 PMCID: PMC3312332 DOI: 10.1155/2012/637452] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2011] [Revised: 11/03/2011] [Accepted: 12/04/2011] [Indexed: 12/14/2022] Open
Abstract
The importance of TNF-alpha in arthritis is well documented. It may be that TNF-alpha is also markedly involved in muscle inflammation (myositis). An animal model where this can be investigated is needed. A newly developed rabbit myositis model involving pronounced muscle overuse and local injections of substances having proinflammatory effects was therefore used in the present study. The aim was to investigate the patterns of TNF-alpha expression in the developing myositis and to evaluate the usefulness of this myositis model for further TNF-alpha research. Human rheumatoid arthritis (RA) synovial tissue was examined as a reference. TNF-alpha immunoexpression and TNF-alpha mRNA, visualized via in situ hybridization, were detected in cells in the inflammatory infiltrates of the affected muscle (soleus muscle). Coexistence of TNF-alpha and CD68 immunoreactions was noted, suggesting that the TNF-alpha reactive cells are macrophages. Expression of TNF-alpha mRNA was also noted in muscle fibers and blood vessel walls in areas with inflammation. These findings demonstrate that TNF-alpha is highly involved in the myositis process. The model can be used in further studies evaluating the importance of TNF-alpha in developing myositis.
Collapse
|
31
|
Dimitri D, Eymard B. Myopathies inflammatoires, myopathies nécrosantes auto-immunes, myopathies génétiques de l’adulte : frontières diagnostiques. Rev Med Interne 2012; 33:134-42. [DOI: 10.1016/j.revmed.2011.11.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 11/26/2011] [Indexed: 10/14/2022]
|
32
|
Morgenroth VH, Hache LP, Clemens PR. Insights into bone health in Duchenne muscular dystrophy. BONEKEY REPORTS 2012; 1:9. [PMID: 23951421 DOI: 10.1038/bonekey.2012.5] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2011] [Accepted: 11/05/2011] [Indexed: 12/22/2022]
Abstract
Poor bone health is a significant problem for patients with Duchenne muscular dystrophy (DMD), a progressive, disabling disease. Although the primary focus of DMD disease pathogenesis is degeneration of striated muscle, impairment of bone health likely has a role in the disease that has only been superficially examined to date. Deficiency of bone mineral density and increased incidence of bone fractures are well-recognized clinical components of the DMD phenotype. Furthermore, therapy with corticosteroids, an approved treatment for DMD that prolongs ambulation, may have multiple effects on bone health in DMD patients. This review examines the evidence in preclinical models and in human DMD disease that provides insight into the role performed by bone in the disease pathogenesis and phenotype of DMD. The information reviewed here points toward the need for mechanistic and therapeutic studies to optimize bone health in DMD patients.
Collapse
|
33
|
Cruz Guzmán ODR, Chávez García AL, Rodríguez-Cruz M. Muscular dystrophies at different ages: metabolic and endocrine alterations. Int J Endocrinol 2012; 2012:485376. [PMID: 22701119 PMCID: PMC3371686 DOI: 10.1155/2012/485376] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2011] [Accepted: 04/02/2012] [Indexed: 12/15/2022] Open
Abstract
Common metabolic and endocrine alterations exist across a wide range of muscular dystrophies. Skeletal muscle plays an important role in glucose metabolism and is a major participant in different signaling pathways. Therefore, its damage may lead to different metabolic disruptions. Two of the most important metabolic alterations in muscular dystrophies may be insulin resistance and obesity. However, only insulin resistance has been demonstrated in myotonic dystrophy. In addition, endocrine disturbances such as hypogonadism, low levels of testosterone, and growth hormone have been reported. This eventually will result in consequences such as growth failure and delayed puberty in the case of childhood dystrophies. Other consequences may be reduced male fertility, reduced spermatogenesis, and oligospermia, both in childhood as well as in adult muscular dystrophies. These facts all suggest that there is a need for better comprehension of metabolic and endocrine implications for muscular dystrophies with the purpose of developing improved clinical treatments and/or improvements in the quality of life of patients with dystrophy. Therefore, the aim of this paper is to describe the current knowledge about of metabolic and endocrine alterations in diverse types of dystrophinopathies, which will be divided into two groups: childhood and adult dystrophies which have different age of onset.
Collapse
Affiliation(s)
- Oriana del Rocío Cruz Guzmán
- Laboratorio de Biología Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, 06703 Ciudad México, DF, Mexico
| | - Ana Laura Chávez García
- Laboratorio de Biología Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, 06703 Ciudad México, DF, Mexico
| | - Maricela Rodríguez-Cruz
- Laboratorio de Biología Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, 06703 Ciudad México, DF, Mexico
- *Maricela Rodríguez-Cruz:
| |
Collapse
|
34
|
Radley-Crabb HG, Fiorotto ML, Grounds MD. The different impact of a high fat diet on dystrophic mdx and control C57Bl/10 mice. PLOS CURRENTS 2011; 3:RRN1276. [PMID: 22094293 PMCID: PMC3217191 DOI: 10.1371/currents.rrn1276] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/24/2011] [Indexed: 01/26/2023]
Abstract
The absence of functional dystrophin protein in patients with Duchenne muscular dystrophy (DMD) and dystrophic mdx mice leads to fragile myofibre membranes and cycles of myofibre necrosis and regeneration. It is proposed that both DMD patients and mdx mice have an altered metabolism and impaired energy status and that nutritional supplementation may reduce the severity of dystropathology. This research compares the in vivo responses of dystrophic mdx and normal control C57Bl/10 mice to a high protein (50%) or a high fat (16%) diet. Consumption of a high protein diet had minimal effects on the body composition or muscle morphology in both strains of mice. In contrast, differences between the strains were seen in response to the high fat diet; this response also varied between mdx mice aged <24 weeks, and mdx mice aged 24 - 40 weeks. C57Bl/10 mice demonstrated many negative side effects after consuming the high fat diet, including weight gain, increased body fat, and elevated inflammatory cytokines. In contrast, after consuming the high fat diet for 16 weeks the mdx mice (< 24 weeks) remained lean with minimal fat deposition and were resistant to changes in body composition. These results support the proposal that energy metabolism in dystrophic mdx mice is altered compared to normal C57Bl/10 mice and this enables the mdx mice to better metabolise the high fat diet and avoid fat deposition. However, older mdx mice (24 - 40-week-old), with increased energy intake, exhibited some mild adverse effects of a high fat diet but to a far lesser extent than age-matched C57Bl/10 mice. Benefits of the high fat diet on dystrophic muscles of young mice were demonstrated by the significantly increased running ability (km) of voluntarily exercised mdx mice and significantly reduced myofibre necrosis in 24-week-old sedentary mdx mice. These novel data clearly identify an 'altered' response to a high fat diet in dystrophic mdx compared to normal C57Bl/10 mice. Our data indicate that the high fat diet may better meet the energy needs of mdx mice to reduce muscle damage and improve muscle function.
Collapse
Affiliation(s)
- Hannah G Radley-Crabb
- School of Anatomy and Human Biology, the University of Western Australia, Perth, Australia and USDA/ARS Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, Texas, USA
| | | | | |
Collapse
|
35
|
Lane RJM, Roncaroli F, Charles P, McGonagle DG, Orrell RW. Acetylcholine receptor antibodies in patients with genetic myopathies: clinical and biological significance. Neuromuscul Disord 2011; 22:122-8. [PMID: 21872471 DOI: 10.1016/j.nmd.2011.06.751] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2011] [Revised: 06/05/2011] [Accepted: 06/27/2011] [Indexed: 10/17/2022]
Abstract
We report two patients with facioscapulohumeral muscular dystrophy (FSHD) presenting with atypical clinical features. Both were found to have antibodies to acetylcholine receptor (AChR-abs) and improved with immunosuppression. AChR-abs have also been reported in patients with other genetic myopathies and it is unlikely that the association is coincidental. There is increasing evidence that muscle fibre degeneration can cause innate immune responses (autoinflammation) that may lead to the breaking of immune tolerance and the generation of autoantibodies to muscle proteins. We compare and contrast this process with the pathogenesis of archetypical myasthenia gravis.
Collapse
Affiliation(s)
- Russell J M Lane
- Charing Cross Hospital, Imperial College London, United Kingdom.
| | | | | | | | | |
Collapse
|
36
|
Radley-Crabb H, Terrill J, Shavlakadze T, Tonkin J, Arthur P, Grounds M. A single 30 min treadmill exercise session is suitable for 'proof-of concept studies' in adult mdx mice: a comparison of the early consequences of two different treadmill protocols. Neuromuscul Disord 2011; 22:170-82. [PMID: 21835619 DOI: 10.1016/j.nmd.2011.07.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2011] [Revised: 06/24/2011] [Accepted: 07/11/2011] [Indexed: 11/15/2022]
Abstract
The extent of muscle pathology in sedentary adult mdx mice is very low and treadmill exercise is often used to increase myofibre necrosis; however, the early events in dystrophic muscle and blood in response to treadmill exercise (leading to myofibre necrosis) are unknown. This study describes in detail two standardised protocols for the treadmill exercise of mdx mice and profiles changes in molecular and cellular events after a single 30 min treadmill session (Protocol A) or after 4 weeks of (twice weekly) treadmill exercise (Protocol B). Both treadmill protocols increased multiple markers of muscle damage. We conclude that a single 30 min treadmill exercise session is a sufficient and conveniently fast screening test and could be used in 'proof-of-concept' studies to evaluate the benefits of pre-clinical drugs in vivo. Myofibre necrosis, blood serum CK and oxidative stress (specifically the ratio of oxidised to reduced protein thiols) are reliable markers of muscle damage after exercise; many parameters demonstrated high biological variation including changes in mRNA levels for key inflammatory cytokines in muscle. The sampling (sacrifice and tissue collection) time after exercise for these parameters is critical. A more precise understanding of the changes in dystrophic muscle after exercise aims to identify biomarkers and new potential therapeutic drug targets for Duchenne Muscular Dystrophy.
Collapse
Affiliation(s)
- Hannah Radley-Crabb
- School of Anatomy and Human Biology, The University of Western Australia, Crawley, Australia.
| | | | | | | | | | | |
Collapse
|
37
|
Dahiya S, Givvimani S, Bhatnagar S, Qipshidze N, Tyagi SC, Kumar A. Osteopontin-stimulated expression of matrix metalloproteinase-9 causes cardiomyopathy in the mdx model of Duchenne muscular dystrophy. THE JOURNAL OF IMMUNOLOGY 2011; 187:2723-31. [PMID: 21810612 DOI: 10.4049/jimmunol.1101342] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, is a common and lethal form of muscular dystrophy. With progressive disease, most patients succumb to death from respiratory or heart failure, or both. However, the mechanisms, especially those governing cardiac inflammation and fibrosis in DMD, remain less understood. Matrix metalloproteinase (MMPs) are a group of extracellular matrix proteases involved in tissue remodeling in both physiologic and pathophysiologic conditions. Previous studies have shown that MMP-9 exacerbates myopathy in dystrophin-deficient mdx mice. However, the role and the mechanisms of action of MMP-9 in cardiac tissue and the biochemical mechanisms leading to increased levels of MMP-9 in mdx mice remain unknown. Our results demonstrate that the levels of MMP-9 are increased in the heart of mdx mice. Genetic ablation of MMP-9 attenuated cardiac injury, left ventricle dilation, and fibrosis in 1-y-old mdx mice. Echocardiography measurements showed improved heart function in Mmp9-deficient mdx mice. Deletion of the Mmp9 gene diminished the activation of ERK1/2 and Akt kinase in the heart of mdx mice. Ablation of MMP-9 also suppressed the expression of MMP-3 and MMP-12 in the heart of mdx mice. Finally, our experiments have revealed that osteopontin, an important immunomodulator, contributes to the increased amounts of MMP-9 in cardiac and skeletal muscle of mdx mice. This study provides a novel mechanism for development of cardiac dysfunction and suggests that MMP-9 and OPN are important therapeutic targets to mitigating cardiac abnormalities in patients with DMD.
Collapse
Affiliation(s)
- Saurabh Dahiya
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | | | | | | | | | | |
Collapse
|
38
|
Matthews E, Miller J, Macleod M, Ironside J, Ambler G, Labrum R, Sud R, Holton J, Hanna M. Sodium and chloride channelopathies with myositis: coincidence or connection? Muscle Nerve 2011; 44:283-8. [PMID: 21698652 PMCID: PMC3136616 DOI: 10.1002/mus.22120] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/25/2010] [Indexed: 11/10/2022]
Abstract
INTRODUCTION A proximal myopathy develops in some patients with muscle channelopathies, but the causative molecular mechanisms are unknown. METHODS We reviewed retrospectively all clinical and muscle biopsy findings of 3 patients with channelopathy and additional myositis. Direct DNA sequencing was performed. RESULTS Pathogenic mutations were identified in each case. Biopsies demonstrated inflammatory infiltrates. CONCLUSIONS Clinicians should consider muscle biopsy in channelopathy patients with severe myalgia and/or subacute weakness and accompanying elevated creatine kinase. Chance association of myositis and channelopathy is statistically unlikely. An alternative hypothesis suggests that inflammatory insults could contribute to myopathy in some patients.
Collapse
Affiliation(s)
- E. Matthews
- MRC Centre for Neuromuscular Disease, UCL, Institute of Neurology, Queen Square, London, WC1N 3BG
| | - J.A.L. Miller
- Department of Neurology, Royal Victoria Infirmary, Newcastle upon Tyne, NE1 4LP
| | - M.R. Macleod
- Department of Neurology, Stirling Royal Infirmary, Stirling, FK8 2AU, UK
- Clinical Neurosciences, University of Edinburgh, Scotland, EH4 2XU, UK
| | - J. Ironside
- Division of Pathology, School of Molecular and Clinical Medicine, University of Edinburgh, Western General Hospital, Edinburgh, EH4 2XU
| | - G. Ambler
- Department of Statistical Science, UCL, 1-19 Torrington Place, London, WC1E 7HB
| | - R. Labrum
- Neurogenetics Unit, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG
| | - R. Sud
- Neurogenetics Unit, National Hospital for Neurology and Neurosurgery, Queen Square, London, WC1N 3BG
| | - J.L. Holton
- MRC Centre for Neuromuscular Disease, UCL, Institute of Neurology, Queen Square, London, WC1N 3BG
| | - M.G. Hanna
- MRC Centre for Neuromuscular Disease, UCL, Institute of Neurology, Queen Square, London, WC1N 3BG
| |
Collapse
|
39
|
Klyen BR, Shavlakadze T, Radley-Crabb HG, Grounds MD, Sampson DD. Identification of muscle necrosis in the mdx mouse model of Duchenne muscular dystrophy using three-dimensional optical coherence tomography. JOURNAL OF BIOMEDICAL OPTICS 2011; 16:076013. [PMID: 21806274 DOI: 10.1117/1.3598842] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
Three-dimensional optical coherence tomography (3D-OCT) was used to image the structure and pathology of skeletal muscle tissue from the treadmill-exercised mdx mouse model of human Duchenne muscular dystrophy. Optical coherence tomography (OCT) images of excised muscle samples were compared with co-registered hematoxylin and eosin-stained and Evans blue dye fluorescence histology. We show, for the first time, structural 3D-OCT images of skeletal muscle dystropathology well correlated with co-located histology. OCT could identify morphological features of interest and necrotic lesions within the muscle tissue samples based on intrinsic optical contrast. These findings demonstrate the utility of 3D-OCT for the evaluation of small-animal skeletal muscle morphology and pathology, particularly for studies of mouse models of muscular dystrophy.
Collapse
Affiliation(s)
- Blake R Klyen
- The University of Western Australia, School of Electrical, Electronic and Computer Engineering, Optical+Biomedical Engineering Laboratory, M018, 35 Stirling Highway, Crawley, Western Australia 6009, Australia.
| | | | | | | | | |
Collapse
|
40
|
Quan D. Muscular dystrophies and neurologic diseases that present as myopathy. Rheum Dis Clin North Am 2011; 37:233-44, vi. [PMID: 21444022 DOI: 10.1016/j.rdc.2011.01.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Chronic muscle weakness is a common complaint among patients seen in rheumatology and neuromuscular specialty clinics. This article focuses on adult-onset muscular dystrophies, select hereditary myopathies, and other neuromuscular conditions that must be distinguished from acquired causes of inflammatory muscle disease such as polymyositis. A few organizing principles help to focus the evaluation and narrow the differential diagnosis.
Collapse
Affiliation(s)
- Dianna Quan
- Electromyography Laboratory, Department of Neurology, University of Colorado Denver, Academic office 1 - MS B-185, 12631 East 17th Avenue, Room 5121, Aurora, CO 80045, USA.
| |
Collapse
|
41
|
Gherardi RK. Pathogenic aspects of dermatomyositis, polymyositis and overlap myositis. Presse Med 2011; 40:e209-18. [PMID: 21376512 DOI: 10.1016/j.lpm.2010.12.013] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 12/17/2010] [Indexed: 12/11/2022] Open
Abstract
Inflammatory myopathies (IMs) often have distinct histopathologic features suggesting humorally mediated involvement of the microcirculation in dermatomyositis (DM), including early capillary deposition of the complement C5b-9 membranolytic attack complex (MAC) and secondary ischaemic changes; and CD8 T-cell-mediated and MHC1-restricted autoimmune attack of myofibers in polymyositis (PM) and inclusion body myositis. Novel insights in these specific diseases include emerging evidence that capillary loss involves whole microvascular units in DM, and that regulatory T-cells strongly protect myofibers from experimental autotoxic attack in PM. However, all IMs do not exhibit pathophysiology-relevant histopathologic features of DM or PM. Autoimmune necrotizing myopathies (AINM) occur in the absence of endomysial inflammatory cells and may be specifically associated with anti-SRP autoantibodies. Moreover, IM histopathological features may be scarce, unspecific and overlapping. Therefore, increasing attention is paid to features shared by IMs regardless of their type, relevant to the innate immune response and to non-immune mechanisms. Innate immune responses to myodamage (and/or as yet unknown stimuli), involves release of chemokines, activation of specific Toll-like receptors (TLRs) and complex Th-1, Th-17 and other cytokine interplays; it triggers DC recruitment and maturation, and is associated with type 1 IFN signature (especially in DM where type 1 IFN-producing cells called plasmacytoid DCs are mainly detected). Non-immune mechanisms mainly include endoplasmic reticulum (ER) stress induced in myofibers by up-regulation of MHC-class I antigens (as typically observed in PM with a diffuse pattern and in DM with perifascicular predominance). ER stress may favour autoimmune reactions but may also be associated with myofiber damage and dysfunction in the absence of lymphocytes. Overlap myositis (OM) may be associated with other connective tissue diseases and a variety of autoantibodies, such as those directed against tRNA synthetase. Myositis specific autoantibodies are mainly expressed by regenerating myofibers, that may also express MHC-1 and endogenous ligand-binding TLRs, thus drawing a picture in which the regenerating myofiber plays a central pathophysiologic role.
Collapse
Affiliation(s)
- Romain K Gherardi
- CHU Henri-Mondor, AP-HP, centre de référence des maladies neuromusculaires Garches-Necker_Mondor-Hendaye, service d'histologie, 94010 Créteil, France.
| |
Collapse
|
42
|
|
43
|
Henningsen J, Pedersen BK, Kratchmarova I. Quantitative analysis of the secretion of the MCP family of chemokines by muscle cells. ACTA ACUST UNITED AC 2011; 7:311-21. [DOI: 10.1039/c0mb00209g] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
44
|
TNF inhibits Notch-1 in skeletal muscle cells by Ezh2 and DNA methylation mediated repression: implications in duchenne muscular dystrophy. PLoS One 2010; 5:e12479. [PMID: 20814569 PMCID: PMC2930001 DOI: 10.1371/journal.pone.0012479] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2009] [Accepted: 08/01/2010] [Indexed: 12/21/2022] Open
Abstract
Background Classical NF-κB signaling functions as a negative regulator of skeletal myogenesis through potentially multiple mechanisms. The inhibitory actions of TNFα on skeletal muscle differentiation are mediated in part through sustained NF-κB activity. In dystrophic muscles, NF-κB activity is compartmentalized to myofibers to inhibit regeneration by limiting the number of myogenic progenitor cells. This regulation coincides with elevated levels of muscle derived TNFα that is also under IKKβ and NF-κB control. Methodology/Principal Findings Based on these findings we speculated that in DMD, TNFα secreted from myotubes inhibits regeneration by directly acting on satellite cells. Analysis of several satellite cell regulators revealed that TNFα is capable of inhibiting Notch-1 in satellite cells and C2C12 myoblasts, which was also found to be dependent on NF-κB. Notch-1 inhibition occurred at the mRNA level suggesting a transcriptional repression mechanism. Unlike its classical mode of action, TNFα stimulated the recruitment of Ezh2 and Dnmt-3b to coordinate histone and DNA methylation, respectively. Dnmt-3b recruitment was dependent on Ezh2. Conclusions/Significance We propose that in dystrophic muscles, elevated levels of TNFα and NF-κB inhibit the regenerative potential of satellite cells via epigenetic silencing of the Notch-1 gene.
Collapse
|
45
|
Treatment of Duchenne muscular dystrophy with ciclosporin A: a randomised, double-blind, placebo-controlled multicentre trial. Lancet Neurol 2010; 9:1053-9. [PMID: 20801085 DOI: 10.1016/s1474-4422(10)70196-4] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
BACKGROUND Duchenne muscular dystrophy is a rare X-linked progressive disease characterised by loss of ambulation at about age 10 years, with death in early adulthood due to respiratory and cardiac insufficiency. Steroids are effective at slowing the progression of muscle weakness; however, their use is limited by side-effects, prompting the search for alternatives. We assessed the effect of ciclosporin A as monotherapy and in combination with intermittent prednisone for the treatment of ambulant patients with this disorder. METHODS Our study was a parallel-group, placebo-controlled, double-blind, multicentre trial at trial sites of the German muscular dystrophy network, MD-NET, over 36 months. Ambulant patients with Duchenne muscular dystrophy who were aged 5 years or older were randomly assigned to receive either ciclosporin A (3·5-4·0 mg/kg per day) or matching placebo. Allocation was done centrally with computer-generated random numbers. Patients and investigators were masked to the allocated treatment. After 3 months of treatment, both groups were also given intermittent prednisone for a further 12 months (0·75 mg/kg, alternating 10 days on with 10 days off). All patients who received at least one dose of study drug or placebo were included in the primary analysis. The primary outcome measure was manual muscle strength measured on the Medical Research Council (MRC) scale. This trial is registered with the German clinical trial register DRKS, number DRKS00000445. FINDINGS 77 patients were randomly assigned to the ciclosporin A group and 76 to the placebo group; 73 patients on ciclosporin A and 73 on placebo received at least one dose and were available for efficacy analyses. 3 months of treatment with ciclosporin A alone did not show any significant improvement in primary outcome measures (mean change in the proportion of a possible total MRC score [%MRC] was -2·6 [SD 6·0] for patients on ciclosporin A and -0·8 [4·9] for patients on placebo; adjusted group difference estimate -0·88, 97·5% CI -2·6 to 0·9; p=0·26). The combination of ciclosporin A with intermittent steroids was not better than intermittent steroids alone over 12 months (mean change in %MRC was 0·7 [7·1] for patients on ciclosporin A and -0·3 [7·9] for patients on placebo; adjusted group difference estimate -0·85, -3·6 to 1·9; p=0·48). Numbers of adverse events (75 in patients on ciclosporin A and 74 on placebo) and serious adverse events (four with ciclosporin A and four with placebo) did not differ significantly between groups. INTERPRETATION Ciclosporin A alone or in combination with intermittent prednisone does not improve muscle strength or functional abilities in ambulant boys with Duchenne muscular dystrophy, but is safe and well tolerated. FUNDING German Federal Ministry of Education and Research, Action Benni and co eV, Novartis Pharma AG, and Deutsche Gesellschaft für Muskelkranke eV.
Collapse
|
46
|
Shi H, Boadu E, Mercan F, Le AM, Flach RJR, Zhang L, Tyner KJ, Olwin BB, Bennett AM. MAP kinase phosphatase-1 deficiency impairs skeletal muscle regeneration and exacerbates muscular dystrophy. FASEB J 2010; 24:2985-97. [PMID: 20371627 DOI: 10.1096/fj.09-150045] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
In skeletal muscle, the mitogen-activated protein kinase (MAPK) phosphatase-1 (MKP-1) is a critical negative regulator of the MAPKs. Since the MAPKs have been reported to be both positive and negative for myogenesis, the physiological role of MKP-1 in skeletal muscle repair and regeneration has remained unclear. Here, we show that MKP-1 plays an essential role in adult regenerative myogenesis. In a cardiotoxin-induced muscle injury model, lack of MKP-1 impaired muscle regeneration. In mdx mice, MKP-1 deficiency reduced body weight, muscle mass, and muscle fiber cross-sectional area. In addition, MKP-1-deficient muscles exhibit exacerbated myopathy accompanied by increased inflammation. Lack of MKP-1 compromised myoblast proliferation and induced precocious differentiation, phenotypes that were rescued by pharmacological inhibition of p38alpha/beta MAPK. MKP-1 coordinates both myoblast proliferation and differentiation. Mechanistically, MyoD bound to the MKP-1 promoter and activated MKP-1 expression in proliferating myoblasts. Later, during myogenesis, MyoD uncoupled from the MKP-1 promoter leading to the down-regulation of MKP-1 and facilitation of promyogenic p38alpha/beta MAPK signaling. Hence, MKP-1 plays a critical role in muscle stem cells and in the immune response to coordinate muscle repair and regeneration.
Collapse
Affiliation(s)
- Hao Shi
- Yale University School of Medicine, Department of Pharmacology, 333 Cedar St., New Haven, CT 06520-8066, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lafoux A, Divet A, Gervier P, Huchet-Cadiou C. Diaphragm tension reduced in dystrophic mice by an oxidant, hypochlorous acid. Can J Physiol Pharmacol 2010; 88:130-40. [PMID: 20237587 DOI: 10.1139/y09-117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
In dystrophin-deficient skeletal muscle cells, in which Ca2+ homeostasis is disrupted and reactive oxygen species production is increased, we hypothesized that hypochlorous acid (HOCl), a strong H2O2-related free radical, damages contractile proteins and the sarcoplasmic reticulum. The aim of the present study was to investigate the effects of exposure to oxidative stress, generated by applying HOCl (100 micromol/L and 1 mmol/L), on the contractile function and sarcoplasmic reticulum properties of dystrophic mice. Experiments were performed on diaphragm muscle, which is severely affected in the mdx mouse, and the results were compared with those obtained in healthy (non-dystrophic) mice. In Triton-skinned fibres from C57BL/10 and mdx mice, 1 mmol/L HOCl increased myofibrillar Ca2+ sensitivity, but decreased maximal Ca2+-activated tension. In the presence of HOCl, higher concentrations of MgATP were required to produce rigor tensions. The interaction between HOCl and the Ca2+ uptake mechanisms was demonstrated using saponin-skinned fibres and sarcoplasmic reticulum vesicles. The results showed that HOCl, at micromolar or millimolar concentrations, can modify sarcoplasmic reticulum Ca2+ uptake and that this effect was more pronounced in diaphragm muscle from mdx mice. We conclude that in dystrophic diaphragm skeletal muscle cells, HOCl activates a cellular pathway that leads to an increase in the intracellular concentration of Ca2+.
Collapse
Affiliation(s)
- Aude Lafoux
- Université de Nantes, CNRS, UMR 6204, Biotechnologie, Biocatalyse et Biorégulation, Faculté des Sciences et des Techniques, 2 rue de la Houssinière, BP 92208, F-44322 Nantes, CEDEX 03, France
| | | | | | | |
Collapse
|
48
|
Brigitte M, Schilte C, Plonquet A, Baba-Amer Y, Henri A, Charlier C, Tajbakhsh S, Albert M, Gherardi RK, Chrétien F. Muscle resident macrophages control the immune cell reaction in a mouse model of notexin-induced myoinjury. ACTA ACUST UNITED AC 2010; 62:268-79. [PMID: 20039420 DOI: 10.1002/art.27183] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE Skeletal muscle may be the site of a variety of poorly understood immune reactions, particularly after myofiber injury, which is typically observed in inflammatory myopathies. This study was undertaken to explore both the cell dynamics and functions of resident macrophages and dendritic cells (DCs) in damaged muscle, using a mouse model of notexin-induced myoinjury to study innate immune cell reactions. METHODS The myeloid cell reaction to notexin-induced myoinjury was analyzed by microscopy and flow cytometry. Bone marrow (BM) transplantation studies were used to discriminate resident from exudate monocyte/macrophages. Functional tests included cytokine screening and an alloantigenic mixed leukocyte reaction to assess the antigen-presenting cell (APC) function. Selective resident macrophage depletion was obtained by injection of diphtheria toxin (DT) into CD11b-DT receptor-transgenic mice transplanted with DT-insensitive BM. RESULTS The connective tissue surrounding mouse muscle/fascicle tissue (the epimysium/perimysium) after deep muscle injury displayed a resident macrophage population of CD11b+F4/80+CD11c-Ly-6C-CX3CR1- cells, which concentrated first in the epimysium. These resident macrophages were being used by leukocytes as a centripetal migration pathway, and were found to selectively release 2 chemokines, cytokine-induced neutrophil chemoattractant and monocyte chemoattractant protein 1, and to crucially contribute to massive recruitment of neutrophils and monocytes from the blood. Early epimysial inflammation consisted of a predominance of Ly-6C(high)CX3CR1(low)CD11c- cells that were progressively substituted by Ly-6C(low)CX3CR1(high) cells displaying an intermediate, rather than high, level of CD11c expression. These CD11c(intermediate) cells were derived from circulating CCR2+ monocytes, functionally behaved as immature APCs in the absence of alloantigenic challenge, and migrated to draining lymph nodes while acquiring the phenotype of mature DCs (CD11c+Ia+CD80+ cells, corresponding to an inflammatory DC phenotype). CONCLUSION The results in this mouse model show that resident macrophages in the muscle epimysium/perimysium orchestrate the innate immune response to myoinjury, which is linked to adaptive immunity through the formation of inflammatory DCs.
Collapse
Affiliation(s)
- Madly Brigitte
- INSERM U955, Groupe Hospitalier Henri Mondor-Albert Chenevier, Assistance Publique Hôpitaux de Paris, Université Paris 12 Val-de-Marne, Créteil, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Angelucci F, Colantoni L. Facioscapulohumeral muscular dystrophy: do neurotrophins play a role? Muscle Nerve 2010; 41:120-7. [PMID: 19813193 DOI: 10.1002/mus.21505] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Although the molecular defect of facioscapulohumeral muscular dystrophy (FSHD) is well established and involves the contraction of the polymorphic 3.3 kb D4Z4 repeat on the subtelomeric region of chromosome 4q35, the pathologic effects of this deletion remain largely unknown. As a consequence, no specific treatment for FSHD is at present available. Thus, there is the need to explore new areas in an attempt to better characterize pathophysiological alterations in FSHD that might be useful for managing the disease. Neurotrophins (nerve growth factor, brain-derived neurotrophic factor, neurotrophin-3, and neurotrophin-4/5) are a class of proteins involved in the development, maintenance, and function of neurons of the peripheral and central nervous systems. In addition, neurotrophins and their RNAs are expressed in muscle, where they have a role in development and regeneration. In this article we put together the experimental evidence that indicates neurotrophins might be involved in the pathophysiology of FSHD and discuss the possible implications of this assumption.
Collapse
Affiliation(s)
- Francesco Angelucci
- IRCCS Santa Lucia Foundation, Department of Clinical and Behavioural Neurology, 00179, Rome, Italy.
| | | |
Collapse
|
50
|
Evans NP, Misyak SA, Robertson JL, Bassaganya-Riera J, Grange RW. Immune-mediated mechanisms potentially regulate the disease time-course of duchenne muscular dystrophy and provide targets for therapeutic intervention. PM R 2009; 1:755-68. [PMID: 19695529 DOI: 10.1016/j.pmrj.2009.04.010] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2009] [Revised: 04/23/2009] [Accepted: 04/28/2009] [Indexed: 11/19/2022]
Abstract
Duchenne muscular dystrophy is a lethal muscle-wasting disease that affects boys. Mutations in the dystrophin gene result in the absence of the dystrophin glycoprotein complex (DGC) from muscle plasma membranes. In healthy muscle fibers, the DGC forms a link between the extracellular matrix and the cytoskeleton to protect against contraction-induced membrane lesions and to regulate cell signaling. The absence of the DGC results in aberrant regulation of inflammatory signaling cascades. Inflammation is a key pathological characteristic of dystrophic muscle lesion formation. However, the role and regulation of this process in the disease time-course has not been sufficiently examined. The transcription factor nuclear factor-kappaB has been shown to contribute to the disease process and is likely involved with increased inflammatory gene expression, including cytokines and chemokines, found in dystrophic muscle. These aberrant signaling processes may regulate the early time-course of inflammatory events that contribute to the onset of disease. This review critically evaluates the possibility that dystrophic muscle lesions in both patients with Duchenne muscular dystrophy and mdx mice are the result of immune-mediated mechanisms that are regulated by inflammatory signaling and also highlights new therapeutic directions.
Collapse
Affiliation(s)
- Nicholas P Evans
- Department of Human Nutrition, Foods and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061-0002, USA.
| | | | | | | | | |
Collapse
|