1
|
Wehling-Henricks M, Kok SY, Gamboa H, Kannan P, Thomas C, Flores I, Welc SS, Tidball JG. Cytotoxic T-Lymphocyte-Associated Protein 4 Fused to a Modified Fragment of IgG1 Reduces Muscle Fiber Damage in a Model of Duchenne Muscular Dystrophy by Attenuating Proinflammatory Gene Expression in Myeloid Lineage Cells. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:717-740. [PMID: 39814100 PMCID: PMC11959425 DOI: 10.1016/j.ajpath.2024.12.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/22/2024] [Accepted: 12/11/2024] [Indexed: 01/18/2025]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, muscle-wasting, genetic disease that is greatly amplified by an immune response to the diseased muscles. The mdx mouse model of DMD was used to test whether the pathology can be reduced by treatment with a cytotoxic T-lymphocyte-associated protein 4 fused to a modified fragment of IgG1 (CTLA4-Ig) fusion protein that blocks costimulatory signals required for activation of T cells. CTLA4-Ig treatment reduced mdx sarcolemma lesions and reduced the numbers of activated T cells, macrophages, and antigen-presenting cells in mdx muscle and reduced macrophage invasion into muscle fibers. In vitro data showed that CTLA4-Ig acts directly on bone marrow cells and macrophages to modify their function and gene expression. CTLA4-Ig treatment of mdx bone marrow cells diminished their mobility and chemotactic response to chemokine ligand-2. Treating mdx macrophages with CTLA4-Ig reduced their cytolysis of muscle cells in vitro. RNA-sequencing analysis of mdx macrophages showed that CTLA4-Ig reduced expression of genes associated with leukocyte chemotaxis, migration, and extravasation; >90% of those affected genes were tumor necrosis factor-α target genes. Comparison of mdx and wild-type macrophages by RNA sequencing showed that 46% of the genes down-regulated by CTLA4-Ig were genes up-regulated in macrophages by the presence of muscular dystrophy in mice. These findings show that CTLA4-Ig is a promising immunotherapeutic for DMD, and many of its beneficial effects may result from direct actions on macrophages that modify their expression of proinflammatory genes.
Collapse
MESH Headings
- Animals
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/immunology
- Muscular Dystrophy, Duchenne/drug therapy
- Mice, Inbred mdx
- Mice
- Myeloid Cells/metabolism
- Myeloid Cells/drug effects
- Myeloid Cells/pathology
- Disease Models, Animal
- Muscle Fibers, Skeletal/pathology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Macrophages/drug effects
- Macrophages/metabolism
- Mice, Inbred C57BL
- Abatacept/pharmacology
- Immunoglobulin G
- Recombinant Fusion Proteins/pharmacology
- CTLA-4 Antigen
- Male
- Inflammation/genetics
- Inflammation/pathology
- Gene Expression Regulation/drug effects
Collapse
Affiliation(s)
| | - Su-Yin Kok
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California
| | - Haley Gamboa
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California
| | - Pranav Kannan
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California; Molecular, Cellular and Integrative Physiology Program, University of California, Los Angeles, California
| | - Connor Thomas
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California
| | - Ivan Flores
- Molecular, Cellular and Integrative Physiology Program, University of California, Los Angeles, California
| | - Steven S Welc
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, Indiana; Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, Indiana
| | - James G Tidball
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California; Molecular, Cellular and Integrative Physiology Program, University of California, Los Angeles, California; Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California.
| |
Collapse
|
2
|
Hernandez-Torres F, Matias-Valiente L, Alzas-Gomez V, Aranega AE. Macrophages in the Context of Muscle Regeneration and Duchenne Muscular Dystrophy. Int J Mol Sci 2024; 25:10393. [PMID: 39408722 PMCID: PMC11477283 DOI: 10.3390/ijms251910393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/15/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Macrophages are essential to muscle regeneration, as they regulate inflammation, carry out phagocytosis, and facilitate tissue repair. These cells exhibit phenotypic switching from pro-inflammatory (M1) to anti-inflammatory (M2) states during muscle repair, influencing myoblast proliferation, differentiation, and myofiber formation. In Duchenne Muscular Dystrophy (DMD), asynchronous muscle injuries disrupt the normal temporal stages of regeneration, leading to fibrosis and failed regeneration. Altered macrophage activity is associated with DMD progression and physiopathology. Gaining insight into the intricate relationship between macrophages and muscle cells is crucial for creating effective therapies aimed at treating this muscle disorder. This review explores the dynamic functions of macrophages in muscle regeneration and their implications in DMD.
Collapse
Affiliation(s)
- Francisco Hernandez-Torres
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain; (L.M.-V.); (V.A.-G.)
| | - Lidia Matias-Valiente
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain; (L.M.-V.); (V.A.-G.)
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, 23071 Jaen, Spain
| | - Virginia Alzas-Gomez
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain; (L.M.-V.); (V.A.-G.)
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, 23071 Jaen, Spain
| | - Amelia Eva Aranega
- Medina Foundation, Technology Park of Health Sciences, 18016 Granada, Spain; (L.M.-V.); (V.A.-G.)
- Department of Experimental Biology, Faculty of Experimental Sciences, University of Jaen, 23071 Jaen, Spain
| |
Collapse
|
3
|
Oliver T, Nguyen NY, Tully CB, McCormack NM, Sun CM, Fiorillo AA, Heier CR. The glucocorticoid receptor acts locally to protect dystrophic muscle and heart during disease. Dis Model Mech 2024; 17:dmm050397. [PMID: 38770680 PMCID: PMC11139035 DOI: 10.1242/dmm.050397] [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: 07/13/2023] [Accepted: 03/28/2024] [Indexed: 05/22/2024] Open
Abstract
Absence of dystrophin results in muscular weakness, chronic inflammation and cardiomyopathy in Duchenne muscular dystrophy (DMD). Pharmacological corticosteroids are the DMD standard of care; however, they have harsh side effects and unclear molecular benefits. It is uncertain whether signaling by physiological corticosteroids and their receptors plays a modifying role in the natural etiology of DMD. Here, we knocked out the glucocorticoid receptor (GR, encoded by Nr3c1) specifically in myofibers and cardiomyocytes within wild-type and mdx52 mice to dissect its role in muscular dystrophy. Double-knockout mice showed significantly worse phenotypes than mdx52 littermate controls in measures of grip strength, hang time, inflammatory pathology and gene expression. In the heart, GR deletion acted additively with dystrophin loss to exacerbate cardiomyopathy, resulting in enlarged hearts, pathological gene expression and systolic dysfunction, consistent with imbalanced mineralocorticoid signaling. The results show that physiological GR functions provide a protective role during muscular dystrophy, directly contrasting its degenerative role in other disease states. These data provide new insights into corticosteroids in disease pathophysiology and establish a new model to investigate cell-autonomous roles of nuclear receptors and mechanisms of pharmacological corticosteroids.
Collapse
MESH Headings
- Animals
- Mice
- Cardiomyopathies/pathology
- Cardiomyopathies/metabolism
- Dystrophin/metabolism
- Dystrophin/genetics
- Dystrophin/deficiency
- Mice, Inbred C57BL
- Mice, Inbred mdx
- Mice, Knockout
- Muscle, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Muscular Dystrophy, Animal/pathology
- Muscular Dystrophy, Animal/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/metabolism
- Myocardium/pathology
- Myocardium/metabolism
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Myocytes, Cardiac/drug effects
- Phenotype
- Receptors, Glucocorticoid/metabolism
Collapse
Affiliation(s)
- Trinitee Oliver
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Biology, Howard University, Washington, DC 20059, USA
- Graduate School of Biomedical Sciences, Cedars-Sinai Medical Center, West Hollywood, CA 90048, USA
| | - Nhu Y. Nguyen
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Molecular Biomedical Sciences, North Carolina State University, Raleigh, NC 27607, USA
| | - Christopher B. Tully
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Nikki M. McCormack
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Christina M. Sun
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
| | - Alyson A. Fiorillo
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC 20037, USA
- Center for Inherited Muscle Research, Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA
| | - Christopher R. Heier
- Center for Genetic Medicine Research, Children's National Hospital, Washington, DC 20010, USA
- Department of Genomics and Precision Medicine, The George Washington University, Washington, DC 20037, USA
- Center for Inherited Muscle Research, Department of Neurology, Virginia Commonwealth University, Richmond, VA 23298, USA
| |
Collapse
|
4
|
Caballero-Sánchez N, Alonso-Alonso S, Nagy L. Regenerative inflammation: When immune cells help to re-build tissues. FEBS J 2024; 291:1597-1614. [PMID: 36440547 PMCID: PMC10225019 DOI: 10.1111/febs.16693] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/29/2022] [Accepted: 11/18/2022] [Indexed: 11/29/2022]
Abstract
Inflammation is an essential immune response critical for responding to infection, injury and maintenance of tissue homeostasis. Upon injury, regenerative inflammation promotes tissue repair by a timed and coordinated infiltration of diverse cell types and the secretion of growth factors, cytokines and lipids mediators. Remarkably, throughout evolution as well as mammalian development, this type of physiological inflammation is highly associated with immunosuppression. For instance, regenerative inflammation is the consequence of an in situ macrophage polarization resulting in a transition from pro-inflammatory to anti-inflammatory/pro-regenerative response. Immune cells are the first responders upon injury, infiltrating the damaged tissue and initiating a pro-inflammatory response depleting cell debris and necrotic cells. After phagocytosis, macrophages undergo multiple coordinated metabolic and transcriptional changes allowing the transition and dictating the initiation of the regenerative phase. Differences between a highly efficient, complete ad integrum tissue repair, such as, acute skeletal muscle injury, and insufficient regenerative inflammation, as the one developing in Duchenne Muscular Dystrophy (DMD), highlight the importance of a coordinated response orchestrated by immune cells. During regenerative inflammation, these cells interact with others and alter the niche, affecting the character of inflammation itself and, therefore, the progression of tissue repair. Comparing acute muscle injury and chronic inflammation in DMD, we review how the same cells and molecules in different numbers, concentration and timing contribute to very different outcomes. Thus, it is important to understand and identify the distinct functions and secreted molecules of macrophages, and potentially other immune cells, during tissue repair, and the contributors to the macrophage switch leveraging this knowledge in treating diseases.
Collapse
Affiliation(s)
- Noemí Caballero-Sánchez
- Doctoral School of Molecular Cell and Immunobiology, Faculty of Medicine, University of Debrecen, Hungary
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
| | - Sergio Alonso-Alonso
- Instituto Oftalmológico Fernández-Vega, Oviedo, Spain
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, Spain
| | - Laszlo Nagy
- Department of Biochemistry and Molecular Biology, Nuclear Receptor Research Laboratory, Faculty of Medicine, University of Debrecen, Hungary
- Departments Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, and Institute for Fundamental Biomedical Research, Johns Hopkins All Children's Hospital, St Petersburg, Florida, USA
| |
Collapse
|
5
|
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
|
6
|
Nitahara-Kasahara Y, Nakayama S, Kimura K, Yamaguchi S, Kakiuchi Y, Nito C, Hayashi M, Nakaishi T, Ueda Y, Okada T. Immunomodulatory amnion-derived mesenchymal stromal cells preserve muscle function in a mouse model of Duchenne muscular dystrophy. Stem Cell Res Ther 2023; 14:108. [PMID: 37106393 PMCID: PMC10142496 DOI: 10.1186/s13287-023-03337-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is an incurable genetic disease characterized by degeneration and necrosis of myofibers, chronic inflammation, and progressive muscle weakness resulting in premature mortality. Immunosuppressive multipotent mesenchymal stromal cell (MSC) therapy could be an option for DMD patients. We focused on amnion-derived mesenchymal stromal cells (AMSCs), a clinically viable cell source owing to their unique characteristics, such as non-invasive isolation, mitotic stability, ethical acceptability, and minimal risk of immune reaction and cancer. We aimed to identify novel immunomodulatory effects of AMSCs on macrophage polarization and their transplantation strategies for the functional recovery of skeletal and cardiac muscles. METHODS We used flow cytometry to analyze the expression of anti-inflammatory M2 macrophage markers on peripheral blood mononuclear cells (PBMCs) co-cultured with human AMSCs (hAMSCs). hAMSCs were intravenously injected into DMD model mice (mdx mice) to assess the safety and efficacy of therapeutic interventions. hAMSC-treated and untreated mdx mice were monitored using blood tests, histological examinations, spontaneous wheel-running activities, grip strength, and echocardiography. RESULTS hAMSCs induced M2 macrophage polarization in PBMCs via prostaglandin E2 production. After repeated systemic hAMSC injections, mdx mice exhibited a transient downregulation of serum creatin kinase. Limited mononuclear cell infiltration and a decreased number of centrally nucleated fibers were indicative of regenerated myofibers following degeneration, suggesting an improved histological appearance of the skeletal muscle of hAMSC-treated mdx mice. Upregulated M2 macrophages and altered cytokine/chemokine expressions were observed in the muscles of hAMSC-treated mdx mice. During long-term experiments, a significant decrease in the grip strength in control mdx mice significantly improved in the hAMSC-treated mdx mice. hAMSC-treated mdx mice maintained running activity and enhanced daily running distance. Notably, the treated mice could run longer distances per minute, indicating high running endurance. Left ventricular function in DMD mice improved in hAMSC-treated mdx mice. CONCLUSIONS Early systemic hAMSC administration in mdx mice ameliorated progressive phenotypes, including pathological inflammation and motor dysfunction, resulting in the long-term improvement of skeletal and cardiac muscle function. The therapeutic effects might be associated with the immunosuppressive properties of hAMSCs via M2 macrophage polarization. This treatment strategy could provide therapeutic benefits to DMD patients.
Collapse
Affiliation(s)
- Yuko Nitahara-Kasahara
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-Ku, Tokyo, 108-8639, Japan.
| | - Soya Nakayama
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Koichi Kimura
- Department of Laboratory Medicine, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Sho Yamaguchi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Yuko Kakiuchi
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-Ku, Tokyo, 108-8639, Japan
| | - Chikako Nito
- Laboratory for Clinical Research, Collaborative Research Center, Nippon Medical School, Tokyo, Japan
| | - Masahiro Hayashi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Tomoyuki Nakaishi
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Yasuyoshi Ueda
- Regenerative Medicine and Cell Therapy Laboratories, Kaneka Corporation, Kobe, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-Ku, Tokyo, 108-8639, Japan.
| |
Collapse
|
7
|
Mahon N, Glennon JC. The Bi-directional Relationship Between Sleep and Inflammation in Muscular Dystrophies: A Narrative Review. Neurosci Biobehav Rev 2023; 150:105116. [PMID: 36870583 DOI: 10.1016/j.neubiorev.2023.105116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 01/31/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Muscular dystrophies vary in presentation and severity, but are associated with profound disability in many people. Although characterised by muscle weakness and wasting, there is also a very high prevalence of sleep problems and disorders which have significant impacts on quality of life in these individuals. There are no curative therapies for muscular dystrophies, with the only options for patients being supportive therapies to aid with symptoms. Therefore, there is an urgent need for new therapeutic targets and a greater understanding of pathogenesis. Inflammation and altered immunity are factors which have prominent roles in some muscular dystrophies and emerging roles in others such as type 1 myotonic dystrophy, signifying a link to pathogenesis. Interestingly, there is also a strong link between inflammation/immunity and sleep. In this review, we will explore this link in the context of muscular dystrophies and how it may influence potential therapeutic targets and interventions.
Collapse
Affiliation(s)
- Niamh Mahon
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Jeffrey C Glennon
- School of Medicine, University College Dublin, Dublin, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Dublin, Ireland
| |
Collapse
|
8
|
Terrill JR, Huchet C, Le Guiner C, Lafoux A, Caudal D, Tulangekar A, Bryson-Richardson RJ, Sztal TE, Grounds MD, Arthur PG. Muscle Pathology in Dystrophic Rats and Zebrafish Is Unresponsive to Taurine Treatment, Compared to the mdx Mouse Model for Duchenne Muscular Dystrophy. Metabolites 2023; 13:metabo13020232. [PMID: 36837851 PMCID: PMC9963000 DOI: 10.3390/metabo13020232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/31/2023] [Accepted: 02/01/2023] [Indexed: 02/09/2023] Open
Abstract
Inflammation and oxidative stress are strongly implicated in the pathology of Duchenne muscular dystrophy (DMD), and the sulphur-containing amino acid taurine ameliorates both and decreases dystropathology in the mdx mouse model for DMD. We therefore further tested taurine as a therapy using dystrophic DMDmdx rats and dmd zebrafish models for DMD that have a more severe dystropathology. However, taurine treatment had little effect on the indices of dystropathology in both these models. While we and others have previously observed a deficiency in taurine in mdx mice, in the current study we show that the rat and zebrafish models had increased taurine content compared with wild-type, and taurine treatment did not increase muscle taurine levels. We therefore hypothesised that endogenous levels of taurine are a key determinate in potential taurine treatment efficacy. Because of this, we felt it important to measure taurine levels in DMD patient plasma samples and showed that in non-ambulant patients (but not in younger patients) there was a deficiency of taurine. These data suggest that taurine homeostasis varies greatly between species and may be influenced by age and disease progression. The potential for taurine to be an effective therapy may depend on such variables.
Collapse
Affiliation(s)
- Jessica R. Terrill
- School of Molecular Sciences, The University of Western Australia, Perth 6009, Australia
- Correspondence:
| | - Corinne Huchet
- TaRGeT Lab, Translational Research for Gene Therapy, INSERM, UMR 1089, Nantes Université, CHU Nantes, 440200 Nantes, France
| | - Caroline Le Guiner
- TaRGeT Lab, Translational Research for Gene Therapy, INSERM, UMR 1089, Nantes Université, CHU Nantes, 440200 Nantes, France
| | - Aude Lafoux
- Therassay Platform, CAPACITES, Nantes Université, 44007 Nantes, France
| | - Dorian Caudal
- Therassay Platform, CAPACITES, Nantes Université, 44007 Nantes, France
| | - Ankita Tulangekar
- School of Biological Sciences, Monash University, Melbourne 3800, Australia
| | | | - Tamar E. Sztal
- School of Biological Sciences, Monash University, Melbourne 3800, Australia
| | - Miranda D. Grounds
- School of Human Sciences, the University of Western Australia, Perth 6009, Australia
| | - Peter G. Arthur
- School of Molecular Sciences, The University of Western Australia, Perth 6009, Australia
| |
Collapse
|
9
|
Valduga AH, Mizobuti DS, Moraes FDSR, Mâncio RD, Moraes LHR, Hermes TDA, Macedo AB, Minatel E. Protection of dystrophic muscle cells using Idebenone correlates with the interplay between calcium, oxidative stress and inflammation. Int J Exp Pathol 2023; 104:4-12. [PMID: 36565155 PMCID: PMC9845605 DOI: 10.1111/iep.12463] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/18/2022] [Accepted: 11/24/2022] [Indexed: 12/25/2022] Open
Abstract
There is strong cross-talk between abnormal intracellular calcium concentration, high levels of reactive oxygen species (ROS) and an exacerbated inflammatory process in the dystrophic muscles of mdx mice, the experimental model of Duchenne muscular dystrophy (DMD). In this study, we investigated effects of Idebenone, a potent anti-oxidant, on oxidative stress markers, the anti-oxidant defence system, intracellular calcium concentrations and the inflammatory process in primary dystrophic muscle cells from mdx mice. Dystrophic muscle cells were treated with Idebenone (0.05 μM) for 24 h. The untreated mdx muscle cells were used as controls. The MTT assay showed that Idebenone did not have a cytotoxic effect on the dystrophic muscle cells. The Idebenone treatment was able to reduce the levels of oxidative stress markers, such as H2 O2 and 4-HNE, as well as decreasing intracellular calcium influx in the dystrophic muscle cells. Regarding Idebenone effects on the anti-oxidant defence system, an up-regulation of catalase levels, glutathione reductase (GR), glutathione peroxidase (GPx) and superoxide dismutase (SOD) activity was observed in the dystrophic muscle cells. In addition, the Idebenone treatment was also associated with reduction in inflammatory molecules, such as nuclear factor kappa-B (NF-κB) and tumour necrosis factor (TNF) in mdx muscle cells. These outcomes supported the use of Idebenone as a protective agent against oxidative stress and related signalling mechanisms involved in dystrophinopathies, such as DMD.
Collapse
Affiliation(s)
- Amanda Harduim Valduga
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Daniela Sayuri Mizobuti
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Fernanda dos Santos Rapucci Moraes
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Rafael Dias Mâncio
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Luis Henrique Rapucci Moraes
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Túlio de Almeida Hermes
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Aline Barbosa Macedo
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| | - Elaine Minatel
- Departamento de Biologia Estrutural e Funcional, Instituto de BiologiaUniversidade Estadual de Campinas (UNICAMP)CampinasSao PauloBrazil
| |
Collapse
|
10
|
Millozzi F, Papait A, Bouché M, Parolini O, Palacios D. Nano-Immunomodulation: A New Strategy for Skeletal Muscle Diseases and Aging? Int J Mol Sci 2023; 24:1175. [PMID: 36674691 PMCID: PMC9862642 DOI: 10.3390/ijms24021175] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/23/2022] [Accepted: 12/24/2022] [Indexed: 01/11/2023] Open
Abstract
The skeletal muscle has a very remarkable ability to regenerate upon injury under physiological conditions; however, this regenerative capacity is strongly diminished in physio-pathological conditions, such as those present in diseased or aged muscles. Many muscular dystrophies (MDs) are characterized by aberrant inflammation due to the deregulation of both the lymphoid and myeloid cell populations and the production of pro-inflammatory cytokines. Pathological inflammation is also observed in old muscles due to a systemic change in the immune system, known as "inflammaging". Immunomodulation represents, therefore, a promising therapeutic opportunity for different skeletal muscle conditions. However, the use of immunomodulatory drugs in the clinics presents several caveats, including their low stability in vivo, the need for high doses to obtain therapeutically relevant effects, and the presence of strong side effects. Within this context, the emerging field of nanomedicine provides the powerful tools needed to control the immune response. Nano-scale materials are currently being explored as biocarriers to release immunomodulatory agents in the damaged tissues, allowing therapeutic doses with limited off-target effects. In addition, the intrinsic immunomodulatory properties of some nanomaterials offer further opportunities for intervention that still need to be systematically explored. Here we exhaustively review the state-of-the-art regarding the use of nano-sized materials to modulate the aberrant immune response that characterizes some physio-pathological muscle conditions, such as MDs or sarcopenia (the age-dependent loss of muscle mass). Based on our learnings from cancer and immune tolerance induction, we also discuss further opportunities, challenges, and limitations of the emerging field of nano-immunomodulation.
Collapse
Affiliation(s)
- Francesco Millozzi
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy
- IRCCS Fondazione Santa Lucia, Via del Fosso di Fiorano, 64, 00143 Rome, Italy
| | - Andrea Papait
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| | - Marina Bouché
- Department of Anatomical, Histological, Forensic Medicine and Orthopaedic Sciences, Section of Histology and Embryology, Sapienza University of Rome, 00161 Rome, Italy
| | - Ornella Parolini
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| | - Daniela Palacios
- Department of Life Sciences and Public Health, Università Cattolica del Sacro Cuore, Largo Vito, 1, 00168 Rome, Italy
- IRCCS Fondazione Policlinico Universitario A. Gemelli IRCCS, Largo Vito, 1, 00168 Rome, Italy
| |
Collapse
|
11
|
Bencze M. Mechanisms of Myofibre Death in Muscular Dystrophies: The Emergence of the Regulated Forms of Necrosis in Myology. Int J Mol Sci 2022; 24:ijms24010362. [PMID: 36613804 PMCID: PMC9820579 DOI: 10.3390/ijms24010362] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/28/2022] Open
Abstract
Myofibre necrosis is a central pathogenic process in muscular dystrophies (MD). As post-lesional regeneration cannot fully compensate for chronic myofibre loss, interstitial tissue accumulates and impairs muscle function. Muscle regeneration has been extensively studied over the last decades, however, the pathway(s) controlling muscle necrosis remains largely unknown. The recent discovery of several regulated cell death (RCD) pathways with necrotic morphology challenged the dogma of necrosis as an uncontrolled process, opening interesting perspectives for many degenerative disorders. In this review, we focus on how cell death affects myofibres in MDs, integrating the latest research in the cell death field, with specific emphasis on Duchenne muscular dystrophy, the best-known and most common hereditary MD. The role of regulated forms of necrosis in myology is still in its infancy but there is increasing evidence that necroptosis, a genetically programmed form of necrosis, is involved in muscle degenerating disorders. The existence of apoptosis in myofibre demise will be questioned, while other forms of non-apoptotic RCDs may also have a role in myonecrosis, illustrating the complexity and possibly the heterogeneity of the cell death pathways in muscle degenerating conditions.
Collapse
Affiliation(s)
- Maximilien Bencze
- “Biology of the Neuromuscular System” Team, Institut Mondor de Recherche Biomédicale (IMRB), University Paris-Est Créteil, INSERM, U955 IMRB, 94010 Créteil, France;
- École Nationale Vétérinaire d’Alfort, IMRB, 94700 Maisons-Alfort, France
| |
Collapse
|
12
|
Characterisation of Progressive Skeletal Muscle Fibrosis in the Mdx Mouse Model of Duchenne Muscular Dystrophy: An In Vivo and In Vitro Study. Int J Mol Sci 2022; 23:ijms23158735. [PMID: 35955872 PMCID: PMC9369129 DOI: 10.3390/ijms23158735] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 07/28/2022] [Accepted: 08/04/2022] [Indexed: 12/12/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a rare genetic disease leading to progressive muscle wasting, respiratory failure, and cardiomyopathy. Although muscle fibrosis represents a DMD hallmark, the organisation of the extracellular matrix and the molecular changes in its turnover are still not fully understood. To define the architectural changes over time in muscle fibrosis, we used an mdx mouse model of DMD and analysed collagen and glycosaminoglycans/proteoglycans content in skeletal muscle sections at different time points during disease progression and in comparison with age-matched controls. Collagen significantly increased particularly in the diaphragm, quadriceps, and gastrocnemius in adult mdx, with fibrosis significantly correlating with muscle degeneration. We also analysed collagen turnover pathways underlying fibrosis development in cultured primary quadriceps-derived fibroblasts. Collagen secretion and matrix metalloproteinases (MMPs) remained unaffected in both young and adult mdx compared to wt fibroblasts, whereas collagen cross-linking and tissue inhibitors of MMP (TIMP) expression significantly increased. We conclude that, in the DMD model we used, fibrosis mostly affects diaphragm and quadriceps with a higher collagen cross-linking and inhibition of MMPs that contribute differently to progressive collagen accumulation during fibrotic remodelling. This study offers a comprehensive histological and molecular characterisation of DMD-associated muscle fibrosis; it may thus provide new targets for tailored therapeutic interventions.
Collapse
|
13
|
Extracellular polysaccharides purified (Polycan) from Aureobasidium pullulans SM‑2001 improves pathophysiology of dystrophin-deficient mdx mice. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00245-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Abstract
Background
Duchenne muscular dystrophy is a hereditary muscular disease involving degeneration (i.e. atrophy and loss of muscle fibres) of skeletal muscles, including the diaphragm, and progressively severe functional decline. A previous study shows Polycan, a type of β-glucan derived from the black yeast Aureobasidium pullulans (SM-2001), promotes osteogenicity and bone loss, and possesses anti-inflammatory activity to induce inflammatory cytokines in human immune and cancer cells.
Objective
In this study, we evaluated changes in exercise load behaviour measurements and changes in muscle-related physiological indicators following oral administration of Polycan in mdx mice, an experimental animal model of Duchenne muscular dystrophy.
Result
In mdx mice, Polycan prevented weight loss and thickness of skeletal muscle. In addition, by monitoring increases in running time of mice on treadmills and performing a grip strength test, we confirmed reduced muscle function was recovered to some extent after administering Polycan to mdx mice. In addition, we confirmed that Polycan significantly altered mRNA expression in a concentration-dependent manner, whereby myogenic transcription factors (MyoD, Myf5 and Myogenin) increased and FoxO3α, MuRF1 and Atrogin-1 decreased. We aimed to investigate the mechanism of action in Polycan on energy metabolism of p-AMPK, SIRT1 and PGC1α with apoptosis expression levels as factors related to signalling pathways. Expression ratios of cleaved-caspase-3/caspase-3 and Bax/Bcl-2 in the Polycan extract-administered group increased compared with the control group.
Conclusion
These results demonstrate that Polycan can improve and protect muscle atrophy by preventing apoptosis via pathway regulation related to myogenic transcription factors and energy metabolism in mdx mice.
Collapse
|
14
|
Sim I, Jang J, Song J, Lee J, Lim H, Lee HJ, Hwang G, Kwon YV, Lee D, Yoon Y. Paeonia lactiflora extract improves the muscle function of mdx mice, an animal model of Duchenne muscular dystrophy, via downregulating the high mobility group box 1 protein. JOURNAL OF ETHNOPHARMACOLOGY 2022; 289:115079. [PMID: 35149132 DOI: 10.1016/j.jep.2022.115079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 12/10/2021] [Accepted: 02/03/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Paeonia lactiflora Pall. is an ethnopharmacological medicine with a long history of human use for treating various inflammatory diseases in many Asian countries. AIM OF THE STUDY Duchenne muscular dystrophy (DMD) is an X-linked degenerative muscle disease affecting 1 in 3500 males and is characterized by severe muscle inflammation and a progressive decline in muscle function. This study aimed to elucidate the effects of an ethanol extract of the root of Paeonia lactiflora Pall. (PL) on the muscle function in the muscular dystrophy X-linked (mdx) mouse, the most commonly used animal model of DMD. MATERIALS AND METHODS Male mdx mice and wild-type controls aged 5 weeks were orally treated with PL for 4 weeks. The corticosteroid prednisolone was used as a comparator drug. Muscle strength and motor coordination were assessed via the grip-strength and rotarod tests, respectively. Muscle damage was evaluated via histological examination and assessment of plasma creatine-kinase activity. Proteomic analyses were conducted to identify the muscle proteins whose levels were significantly affected by PL (ProteomeXchange identifier: PXD028886). Muscle and plasma levels of these proteins, and their corresponding mRNAs were measured using western blotting and ELISA, and quantitative reverse transcription-polymerase chain reaction, respectively. RESULTS The muscle strength and motor coordination of mdx mice were significantly increased by the oral treatment of PL. PL significantly reduced the histological muscle damage and plasma creatine-kinase activity. Proteomic analyses of the muscle showed that PL significantly downregulated the high mobility group box 1 (HMGB1) protein and Toll-like receptor (TLR) 4, thus suppressing the HMGB1-TLR4-NF-κB signaling, in the muscle of mdx mice. Consequently, the muscle levels of proinflammatory cytokines/chemokines, which play crucial roles in inflammation, were downregulated. CONCLUSION PL improves the muscle function and reduces the muscle damage in mdx mice via suppressing the HMGB1-TLR4-NF-κB signaling and downregulating proinflammatory cytokines/chemokines.
Collapse
Affiliation(s)
- Inae Sim
- Department of Microbiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| | - Jaewoong Jang
- Department of Microbiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| | - Jaewon Song
- Department of Microbiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| | - Jongkyu Lee
- Department of Microbiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| | - Hyemi Lim
- Department of Microbiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| | - Hyun Jung Lee
- Department of Anatomy and Cell Biology, College of Medicine, Chung-Ang University, Seoul, Republic of Korea.
| | - Gyusik Hwang
- Research Center, EBIOGEN Inc., #405, Sungsu A1 Center, 48 Ttukseom-ro 17-ga-gil, Seongdong-gu, Seoul, 04785, Republic of Korea.
| | - Young V Kwon
- Department of Biochemistry, University of Washington, 1959 NE Pacific St, Seattle, WA, 98195, USA.
| | - Doheon Lee
- Department of Bio and Brain Engineering, KAIST, 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Bio-Synergy Research Center, 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| | - Yoosik Yoon
- Department of Microbiology, College of Medicine, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea; Bio-Synergy Research Center, 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
15
|
Singh S, Singh T, Kunja C, Dhoat NS, Dhania NK. Gene-editing, immunological and iPSCs based therapeutics for muscular dystrophy. Eur J Pharmacol 2021; 912:174568. [PMID: 34656607 DOI: 10.1016/j.ejphar.2021.174568] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/25/2021] [Accepted: 10/11/2021] [Indexed: 10/20/2022]
Abstract
Muscular dystrophy is a well-known genetically heterogeneous group of rare muscle disorders. This progressive disease causes the breakdown of skeletal muscles over time and leads to grave weakness. This breakdown is caused by a diverse pattern of mutations in dystrophin and dystrophin associated protein complex. These mutations lead to the production of altered proteins in response to which, the body stimulates production of various cytokines and immune cells, particularly reactive oxygen species and NFκB. Immune cells display/exhibit a dual role by inducing muscle damage and muscle repair. Various anti-oxidants, anti-inflammatory and glucocorticoid drugs serve as potent therapeutics for muscular dystrophy. Along with the above mentioned therapeutics, induced pluripotent stem cells also serve as a novel approach paving a way for personalized treatment. These pluripotent stem cells allow regeneration of large numbers of regenerative myogenic progenitors that can be administered in muscular dystrophy patients which assist in the recovery of lost muscle fibers. In this review, we have summarized gene-editing, immunological and induced pluripotent stem cell based therapeutics for muscular dystrophy treatment.
Collapse
Affiliation(s)
- Shagun Singh
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India
| | - Tejpal Singh
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India
| | - Chaitanya Kunja
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India
| | - Navdeep S Dhoat
- Department of Pediatrics Surgery, All India Institute of Medical Sciences, Bathinda, 151001, Punjab, India
| | - Narender K Dhania
- Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda-151001, Punjab, India.
| |
Collapse
|
16
|
Yao S, Chen Z, Yu Y, Zhang N, Jiang H, Zhang G, Zhang Z, Zhang B. Current Pharmacological Strategies for Duchenne Muscular Dystrophy. Front Cell Dev Biol 2021; 9:689533. [PMID: 34490244 PMCID: PMC8417245 DOI: 10.3389/fcell.2021.689533] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/23/2021] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal, X-linked neuromuscular disorder caused by the absence of dystrophin protein, which is essential for muscle fiber integrity. Loss of dystrophin protein leads to recurrent myofiber damage, chronic inflammation, progressive fibrosis, and dysfunction of muscle stem cells. There is still no cure for DMD so far and the standard of care is principally limited to symptom relief through glucocorticoids treatments. Current therapeutic strategies could be divided into two lines. Dystrophin-targeted therapeutic strategies that aim at restoring the expression and/or function of dystrophin, including gene-based, cell-based and protein replacement therapies. The other line of therapeutic strategies aims to improve muscle function and quality by targeting the downstream pathological changes, including inflammation, fibrosis, and muscle atrophy. This review introduces the important developments in these two lines of strategies, especially those that have entered the clinical phase and/or have great potential for clinical translation. The rationale and efficacy of each agent in pre-clinical or clinical studies are presented. Furthermore, a meta-analysis of gene profiling in DMD patients has been performed to understand the molecular mechanisms of DMD.
Collapse
Affiliation(s)
- Shanshan Yao
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Zihao Chen
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Yuanyuan Yu
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Ning Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Hewen Jiang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Ge Zhang
- Law Sau Fai Institute for Advancing Translational Medicine in Bone and Joint Diseases, School of Chinese Medicine, Hong Kong Baptist University, Kowloon, Hong Kong
| | - Zongkang Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Baoting Zhang
- School of Chinese Medicine, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
17
|
A Blood Biomarker for Duchenne Muscular Dystrophy Shows That Oxidation State of Albumin Correlates with Protein Oxidation and Damage in Mdx Muscle. Antioxidants (Basel) 2021; 10:antiox10081241. [PMID: 34439489 PMCID: PMC8389308 DOI: 10.3390/antiox10081241] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/14/2021] [Accepted: 07/29/2021] [Indexed: 12/31/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe X-linked muscle wasting disease with no cure. While the precise mechanisms of progressive dystropathology remain unclear, oxidative stress caused by excessive generation of oxidants is strongly implicated. Blood biomarkers that could track oxidant levels in tissues would be valuable to measure the effectiveness of clinical treatments for DMD; our research has focused on developing such biomarkers. One target of oxidants that has the potential to be harnessed as a clinical biomarker is the thiol side chain of cysteine 34 (Cys34) of the blood protein albumin. This study using the mdx mouse model of DMD shows that in plasma, albumin Cys34 undergoes thiol oxidation and these changes correlate with levels of protein thiol oxidation and damage of the dystrophic muscles. A comparison with the commonly used biomarker protein carbonylation, confirmed that albumin thiol oxidation is the more sensitive plasma biomarker of oxidative stress occurring in muscle tissue. We show that plasma albumin oxidation reflects muscle dystropathology, as increased after exercise and decreased after taurine treatment of mdx mice. These data support the use of albumin thiol oxidation as a blood biomarker of dystropathology to assist with advancing clinical development of therapies for DMD.
Collapse
|
18
|
Rossi R, Falzarano MS, Osman H, Armaroli A, Scotton C, Mantuano P, Boccanegra B, Cappellari O, Schwartz E, Yuryev A, Mercuri E, Bertini E, D'Amico A, Mora M, Johansson C, Al-Khalili Szigyarto C, De Luca A, Ferlini A. Circadian Genes as Exploratory Biomarkers in DMD: Results From Both the mdx Mouse Model and Patients. Front Physiol 2021; 12:678974. [PMID: 34305639 PMCID: PMC8300012 DOI: 10.3389/fphys.2021.678974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 06/14/2021] [Indexed: 11/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a rare genetic disease due to dystrophin gene mutations which cause progressive weakness and muscle wasting. Circadian rhythm coordinates biological processes with the 24-h cycle and it plays a key role in maintaining muscle functions, both in animal models and in humans. We explored expression profiles of circadian circuit master genes both in Duchenne muscular dystrophy skeletal muscle and in its animal model, the mdx mouse. We designed a customized, mouse-specific Fluidic-Card-TaqMan-based assay (Fluid-CIRC) containing thirty-two genes related to circadian rhythm and muscle regeneration and analyzed gastrocnemius and tibialis anterior muscles from both unexercised and exercised mdx mice. Based on this first analysis, we prioritized the 7 most deregulated genes in mdx mice and tested their expression in skeletal muscle biopsies from 10 Duchenne patients. We found that CSNK1E, SIRT1, and MYOG are upregulated in DMD patient biopsies, consistent with the mdx data. We also demonstrated that their proteins are detectable and measurable in the DMD patients’ plasma. We suggest that CSNK1E, SIRT1, and MYOG might represent exploratory circadian biomarkers in DMD.
Collapse
Affiliation(s)
- Rachele Rossi
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,The Dubowitz Neuromuscular Centre, Institute of Child Health, London, United Kingdom
| | - Maria Sofia Falzarano
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Hana Osman
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,Department of Medical Microbiology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, Sudan
| | - Annarita Armaroli
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Chiara Scotton
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy
| | - Paola Mantuano
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Brigida Boccanegra
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Ornella Cappellari
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | | | | | - Eugenio Mercuri
- Pediatric Neurology Unit, Catholic University and Nemo Center, Policlinico Universitario Gemelli, Rome, Italy
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, IRCCS Bambino Gesu Children's Hospital, Rome, Italy
| | - Adele D'Amico
- Unit of Neuromuscular and Neurodegenerative Disorders, Department of Neurosciences, IRCCS Bambino Gesu Children's Hospital, Rome, Italy
| | - Marina Mora
- Neuromuscular Diseases and Neuroimmunology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Camilla Johansson
- School of Chemistry, Biotechnology and Health, Royal Institute of Technology, Stockholm, Sweden
| | - Cristina Al-Khalili Szigyarto
- School of Chemistry, Biotechnology and Health, Royal Institute of Technology, Stockholm, Sweden.,Science for Life Laboratory, Royal Institute of Technology, Stockholm, Sweden
| | - Annamaria De Luca
- Section of Pharmacology, Department of Pharmacy-Drug Sciences, University of Bari "Aldo Moro", Bari, Italy
| | - Alessandra Ferlini
- Unit of Medical Genetics, Department of Medical Sciences, University of Ferrara, Ferrara, Italy.,The Dubowitz Neuromuscular Centre, Institute of Child Health, London, United Kingdom
| |
Collapse
|
19
|
The Interplay of Mitophagy and Inflammation in Duchenne Muscular Dystrophy. Life (Basel) 2021; 11:life11070648. [PMID: 34357020 PMCID: PMC8307817 DOI: 10.3390/life11070648] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/30/2021] [Accepted: 07/02/2021] [Indexed: 12/11/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked neuromuscular disease caused by a pathogenic disruption of the DYSTROPHIN gene that results in non-functional dystrophin protein. DMD patients experience loss of ambulation, cardiac arrhythmia, metabolic syndrome, and respiratory failure. At the molecular level, the lack of dystrophin in the muscle results in myofiber death, fibrotic infiltration, and mitochondrial dysfunction. There is no cure for DMD, although dystrophin-replacement gene therapies and exon-skipping approaches are being pursued in clinical trials. Mitochondrial dysfunction is one of the first cellular changes seen in DMD myofibers, occurring prior to muscle disease onset and progresses with disease severity. This is seen by reduced mitochondrial function, abnormal mitochondrial morphology and impaired mitophagy (degradation of damaged mitochondria). Dysfunctional mitochondria release high levels of reactive oxygen species (ROS), which can activate pro-inflammatory pathways such as IL-1β and IL-6. Impaired mitophagy in DMD results in increased inflammation and further aggravates disease pathology, evidenced by increased muscle damage and increased fibrosis. This review will focus on the critical interplay between mitophagy and inflammation in Duchenne muscular dystrophy as a pathological mechanism, as well as describe both candidate and established therapeutic targets that regulate these pathways.
Collapse
|
20
|
Chen Y, Ma Y, Feng JJ, Wang YH, Li TF, Nurmi K, Eklund KK, Wen JG. Histamine H 3 Receptor Signaling Regulates the NLRP3 Inflammasome Activation in C2C12 Myocyte During Myogenic Differentiation. Front Pharmacol 2021; 12:599393. [PMID: 34135750 PMCID: PMC8202077 DOI: 10.3389/fphar.2021.599393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 05/05/2021] [Indexed: 11/13/2022] Open
Abstract
NLRP3 inflammasome has been implicated in impaired post-injury muscle healing and in muscle atrophy. Histamine receptors play an important role in inflammation, but the role of histamine H3 receptor (H3R) in myocyte regeneration and in the regulation of NLRP3 inflammasome is not known. We studied the effects of H3R signaling on C2C12 myocyte viability, apoptosis, and tumor necrosis factor alpha (TNFα)-induced NLRP3 inflammasome activation during striated myogenic differentiation at three time points (days 0, 3, and 6). Expression of Nlrp3, interleukin-1β (IL-1β), and myogenesis markers were determined. TNFα reduced overall viability of C2C12 cells, and exposure to TNFα induced apoptosis of cells at D6. Activation of H3R had no effect on viability or apoptosis, whereas inhibition of H3R increased TNFα-induced apoptosis. Stimulation of C2C12 cells with TNFα increased Nlrp3 mRNA expression at D3 and D6. Moreover, TNFα reduced the expression of myogenesis markers MyoD1, Myogenin, and Myosin-2 at D3 and D6. H3R attenuated TNFα-induced expression of Nlrp3 and further inhibited the myogenesis marker expression; while H3R -blockage enhanced the proinflammatory effects of TNFα and increased the myogenesis marker expression. TNFα-induced secretion of mature IL-1β was dependent on the activation of the NLRP3 inflammasome, as shown by the reduced secretion of mature IL-1β upon treatment of the cells with the small molecule inhibitor of the NLRP3 inflammasome (MCC950). The activation of H3R reduced TNFα-induced IL-1β secretion, while the H3R blockage had an opposite effect. In conclusion, the modulation of H3R activity regulates the effects of TNFα on C2C12 myocyte differentiation and TNFα-induced activation of NLRP3 inflammasome. Thus, H3R signaling may represent a novel target for limiting postinjury muscle inflammation and muscle atrophy.
Collapse
Affiliation(s)
- Yan Chen
- Urodynamic Center and Department of Urology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.,Helsinki Rheumatic Diseases and Inflammation Research Group, Clinicum Helsinki University, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki University Clinicum, Helsinki, Finland
| | - Yuan Ma
- Urodynamic Center and Department of Urology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jin Jin Feng
- Urodynamic Center and Department of Urology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yi He Wang
- Urodynamic Center and Department of Urology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Tian Fang Li
- Department of Rheumatology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Katariina Nurmi
- Helsinki Rheumatic Diseases and Inflammation Research Group, Clinicum Helsinki University, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki University Clinicum, Helsinki, Finland
| | - Kari K Eklund
- Helsinki Rheumatic Diseases and Inflammation Research Group, Clinicum Helsinki University, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki University Clinicum, Helsinki, Finland.,Department of Medicine, Division of Rheumatology, Helsinki University Central Hospital, and Orton Orthopedic Hospital, Helsinki, Finland
| | - Jian Guo Wen
- Urodynamic Center and Department of Urology, Institute of Clinical Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Terrill JR, Webb SM, Arthur PG, Hackett MJ. Investigation of the effect of taurine supplementation on muscle taurine content in the mdx mouse model of Duchenne muscular dystrophy using chemically specific synchrotron imaging. Analyst 2021; 145:7242-7251. [PMID: 32893271 DOI: 10.1039/d0an00642d] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal genetic muscle wasting disorder, which currently has no cure. Supplementation with the drug taurine has been shown to offer therapeutic benefit in the mdx model for DMD, however the mechanism by which taurine protects dystrophic muscle is not fully understood. Mdx muscle is deficient in taurine, however it is not known if this deficiency occurs in the extracellular space, in other cells present in the tissue (such as immune cells) or in the myofibre itself. Likewise, the tissue location of taurine enrichment in taurine treated mdx muscle is not known. In this study we applied X-ray absorption near edge spectroscopy (XANES) at the sulfur K-edge in an imaging format to determine taurine distribution in muscle tissue. XANES is the only technique currently capable of imaging taurine directly in muscle tissue, at a spatial resolution approaching myocyte cell size (20-50 μm). Using a multi-modal approach of XANES imaging and histology on the same tissue sections, we show that in mdx muscle, it is the myofibres that are deficient in taurine, and taurine supplementation ameliorates this deficiency. Increasing the taurine content of mdx myofibres was associated with a decrease in myofibre damage (as shown by the percentage of intact myofibres) and inflammation. These data will help drive future studies to better elucidate the molecular mechanisms through which taurine protects dystrophic muscle; they also support the continued investigation of taurine as a therapeutic intervention for DMD.
Collapse
Affiliation(s)
- Jessica R Terrill
- School of Molecular Sciences, the University of Western Australia, Perth, Western Australia AUS 6009, Australia
| | | | | | | |
Collapse
|
22
|
Ziemkiewicz N, Hilliard G, Pullen NA, Garg K. The Role of Innate and Adaptive Immune Cells in Skeletal Muscle Regeneration. Int J Mol Sci 2021; 22:3265. [PMID: 33806895 PMCID: PMC8005179 DOI: 10.3390/ijms22063265] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 03/17/2021] [Accepted: 03/18/2021] [Indexed: 02/06/2023] Open
Abstract
Skeletal muscle regeneration is highly dependent on the inflammatory response. A wide variety of innate and adaptive immune cells orchestrate the complex process of muscle repair. This review provides information about the various types of immune cells and biomolecules that have been shown to mediate muscle regeneration following injury and degenerative diseases. Recently developed cell and drug-based immunomodulatory strategies are highlighted. An improved understanding of the immune response to injured and diseased skeletal muscle will be essential for the development of therapeutic strategies.
Collapse
Affiliation(s)
- Natalia Ziemkiewicz
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, 3507 Lindell Blvd, St. Louis, MO 63103, USA;
| | - Genevieve Hilliard
- Department of Biology, Saint Louis University, St. Louis, MO 63103, USA;
| | - Nicholas A. Pullen
- School of Biological Sciences, College of Natural and Health Sciences, University of Northern Colorado, Greeley, Colorado, CO 80639, USA;
| | - Koyal Garg
- Department of Biomedical Engineering, Parks College of Engineering, Aviation, and Technology, Saint Louis University, 3507 Lindell Blvd, St. Louis, MO 63103, USA;
| |
Collapse
|
23
|
Nitahara-Kasahara Y, Kuraoka M, Guillermo PH, Hayashita-Kinoh H, Maruoka Y, Nakamura-Takahasi A, Kimura K, Takeda S, Okada T. Dental pulp stem cells can improve muscle dysfunction in animal models of Duchenne muscular dystrophy. Stem Cell Res Ther 2021; 12:78. [PMID: 33494794 PMCID: PMC7831244 DOI: 10.1186/s13287-020-02099-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 12/13/2020] [Indexed: 12/11/2022] Open
Abstract
Background Duchenne muscular dystrophy (DMD) is an inherited progressive disorder that causes skeletal and cardiac muscle deterioration with chronic inflammation. Dental pulp stem cells (DPSCs) are attractive candidates for cell-based strategies for DMD because of their immunosuppressive properties. Therefore, we hypothesized that systemic treatment with DPSCs might show therapeutic benefits as an anti-inflammatory therapy. Methods To investigate the potential benefits of DPSC transplantation for DMD, we examined disease progression in a DMD animal model, mdx mice, by comparing them with different systemic treatment conditions. The DPSC-treated model, a canine X-linked muscular dystrophy model in Japan (CXMDJ), which has a severe phenotype similar to that of DMD patients, also underwent comprehensive analysis, including histopathological findings, muscle function, and locomotor activity. Results We demonstrated a therapeutic strategy for long-term functional recovery in DMD using repeated DPSC administration. DPSC-treated mdx mice and CXMDJ showed no serious adverse events. MRI findings and muscle histology suggested that DPSC treatment downregulated severe inflammation in DMD muscles and demonstrated a milder phenotype after DPSC treatment. DPSC-treated models showed increased recovery in grip-hand strength and improved tetanic force and home cage activity. Interestingly, maintenance of long-term running capability and stabilized cardiac function was also observed in 1-year-old DPSC-treated CXMDJ. Conclusions We developed a novel strategy for the safe and effective transplantation of DPSCs for DMD recovery, which included repeated systemic injection to regulate inflammation at a young age. This is the first report on the efficacy of a systemic DPSC treatment, from which we can propose that DPSCs may play an important role in delaying the DMD disease phenotype.
Collapse
Affiliation(s)
- Yuko Nitahara-Kasahara
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan. .,Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo-city, Tokyo, Japan. .,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.
| | - Mutsuki Kuraoka
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Laboratory of Experimental Animal Science, Nippon Veterinary and Life Science University, Musashino, Tokyo, Japan
| | - Posadas Herrera Guillermo
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan.,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan
| | - Hiromi Hayashita-Kinoh
- Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo-city, Tokyo, Japan.,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan
| | - Yasunobu Maruoka
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | | | - Koichi Kimura
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan.,Department of General Medicine, The Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan
| | - Takashi Okada
- Division of Cell and Gene Therapy, Nippon Medical School, Bunkyo-city, Tokyo, Japan. .,Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Tokyo, Japan. .,Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, Institute of Medical Science, The University of Tokyo, Minato-city, Tokyo, Japan.
| |
Collapse
|
24
|
van Westering TLE, Johansson HJ, Hanson B, Coenen-Stass AML, Lomonosova Y, Tanihata J, Motohashi N, Yokota T, Takeda S, Lehtiö J, Wood MJA, El Andaloussi S, Aoki Y, Roberts TC. Mutation-independent Proteomic Signatures of Pathological Progression in Murine Models of Duchenne Muscular Dystrophy. Mol Cell Proteomics 2020; 19:2047-2068. [PMID: 32994316 PMCID: PMC7710136 DOI: 10.1074/mcp.ra120.002345] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Indexed: 12/23/2022] Open
Abstract
The absence of the dystrophin protein in Duchenne muscular dystrophy (DMD) results in myofiber fragility and a plethora of downstream secondary pathologies. Although a variety of experimental therapies are in development, achieving effective treatments for DMD remains exceptionally challenging, not least because the pathological consequences of dystrophin loss are incompletely understood. Here we have performed proteome profiling in tibialis anterior muscles from two murine DMD models (mdx and mdx52) at three ages (8, 16, and 80 weeks of age), all n = 3. High-resolution isoelectric focusing liquid chromatography-tandem MS (HiRIEF-LC-MS/MS) was used to quantify the expression of 4974 proteins across all 27 samples. The two dystrophic models were found to be highly similar, whereas multiple proteins were differentially expressed relative to WT (C57BL/6) controls at each age. Furthermore, 1795 proteins were differentially expressed when samples were pooled across ages and dystrophic strains. These included numerous proteins associated with the extracellular matrix and muscle function that have not been reported previously. Pathway analysis revealed multiple perturbed pathways and predicted upstream regulators, which together are indicative of cross-talk between inflammatory, metabolic, and muscle growth pathways (e.g. TNF, INFγ, NF-κB, SIRT1, AMPK, PGC-1α, PPARs, ILK, and AKT/PI3K). Upregulation of CAV3, MVP and PAK1 protein expression was validated in dystrophic muscle by Western blot. Furthermore, MVP was upregulated during, but not required for, the differentiation of C2C12 myoblasts suggesting that this protein may affect muscle regeneration. This study provides novel insights into mutation-independent proteomic signatures characteristic of the dystrophic phenotype and its progression with aging.
Collapse
Affiliation(s)
| | - Henrik J Johansson
- Department of Oncology/Pathology, Cancer Proteomics Mass Spectrometry, SciLifeLab Stockholm, Karolinska Institutet, Stockholm, Sweden
| | - Britt Hanson
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK
| | | | - Yulia Lomonosova
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - Jun Tanihata
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, Japan
| | - Norio Motohashi
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, Japan
| | - Toshifumi Yokota
- Department of Medical, Genetics, School of Human Development Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Shin'ichi Takeda
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, Japan
| | - Janne Lehtiö
- Department of Oncology/Pathology, Cancer Proteomics Mass Spectrometry, SciLifeLab Stockholm, Karolinska Institutet, Stockholm, Sweden
| | - Matthew J A Wood
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK; MDUK Oxford Neuromuscular Centre, Oxford, UK
| | | | - Yoshitsugu Aoki
- Department of Molecular Therapy, National Institute of Neuroscience, National Center of Neurology and Psychiatry (NCNP), Kodaira, Tokyo, Japan.
| | - Thomas C Roberts
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK; Department of Paediatrics, University of Oxford, Oxford, UK; Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden.
| |
Collapse
|
25
|
Zschüntzsch J, Jouvenal PV, Zhang Y, Klinker F, Tiburcy M, Liebetanz D, Malzahn D, Brinkmeier H, Schmidt J. Long-term human IgG treatment improves heart and muscle function in a mouse model of Duchenne muscular dystrophy. J Cachexia Sarcopenia Muscle 2020; 11:1018-1031. [PMID: 32436338 PMCID: PMC7432639 DOI: 10.1002/jcsm.12569] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 02/10/2020] [Accepted: 02/25/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD) is a progressive muscle-wasting disease caused by mutations in the dystrophin gene, which leads to structural instability of the dystrophin-glycoprotein-complex with subsequent muscle degeneration. In addition, muscle inflammation has been implicated in disease progression and therapeutically addressed with glucocorticosteroids. These have numerous adverse effects. Treatment with human immunoglobulin G (IgG) improved clinical and para-clinical parameters in the early disease phase in the well-established mdx mouse model. The aim of the present study was to confirm the efficacy of IgG in a long-term pre-clinical study in mdx mice. METHODS IgG (2 g/kg body weight) or NaCl solution as control was administered monthly over 18 months by intraperitoneal injection in mdx mice beginning at 3 weeks of age. Several clinical outcome measures including endurance, muscle strength, and echocardiography were assessed. After 18 months, the animals were sacrificed, blood was collected for analysis, and muscle samples were obtained for ex vivo muscle contraction tests, quantitative PCR, and histology. RESULTS IgG significantly improved the daily voluntary running performance (1.9 m more total daily running distance, P < 0.0001) and slowed the decrease in grip strength by 0.1 mN, (P = 0.018). IgG reduced fatigability of the diaphragm (improved ratio to maximum force by 0.09 ± 0.04, P = 0.044), but specific tetanic force remained unchanged in the ex vivo muscle contraction test. Cardiac function was significantly better after IgG, especially fractional area shortening (P = 0.012). These results were accompanied by a reduction in cardiac fibrosis and the infiltration of T cells (P = 0.0002) and macrophages (P = 0.0027). In addition, treatment with IgG resulted in a significant reduction of the infiltration of T cells (P ≤ 0.036) in the diaphragm, gastrocnemius, quadriceps, and a similar trend in tibialis anterior and macrophages (P ≤ 0.045) in gastrocnemius, quadriceps, tibialis anterior, and a similar trend in the diaphragm, as well as a decrease in myopathic changes as reflected by a reduced central nuclear index in the diaphragm, tibialis anterior, and quadriceps (P ≤ 0.002 in all). CONCLUSIONS The present study underscores the importance of an inflammatory contribution to the disease progression of DMD. The data demonstrate the long-term efficacy of IgG in the mdx mouse. IgG is well tolerated by humans and could preferentially complement gene therapy in DMD. The data call for a clinical trial with IgG in DMD.
Collapse
Affiliation(s)
- Jana Zschüntzsch
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Pia Vanessa Jouvenal
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| | - Yaxin Zhang
- Institute of Pathophysiology, University Medicine Greifswald, Karlsburg, Germany
| | - Florian Klinker
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, Göttingen, Germany
| | - David Liebetanz
- Department of Clinical Neurophysiology, University Medical Center Göttingen, Göttingen, Germany
| | - Dörthe Malzahn
- Department of Genetic Epidemiology, University Medical Center Göttingen, Göttingen, Germany.,mzBiostatistics, Statistical Consultancy, Göttingen, Germany
| | - Heinrich Brinkmeier
- Institute of Pathophysiology, University Medicine Greifswald, Karlsburg, Germany
| | - Jens Schmidt
- Department of Neurology, University Medical Center Göttingen, Göttingen, Germany
| |
Collapse
|
26
|
Hightower RM, Reid AL, Gibbs DE, Wang Y, Widrick JJ, Kunkel LM, Kastenschmidt JM, Villalta SA, van Groen T, Chang H, Gornisiewicz S, Landesman Y, Tamir S, Alexander MS. The SINE Compound KPT-350 Blocks Dystrophic Pathologies in DMD Zebrafish and Mice. Mol Ther 2020; 28:189-201. [PMID: 31628052 PMCID: PMC6952030 DOI: 10.1016/j.ymthe.2019.08.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/23/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked muscle wasting disease that is caused by the loss of functional dystrophin protein in cardiac and skeletal muscles. DMD patient muscles become weakened, leading to eventual myofiber breakdown and replacement with fibrotic and adipose tissues. Inflammation drives the pathogenic processes through releasing inflammatory cytokines and other factors that promote skeletal muscle degeneration and contributing to the loss of motor function. Selective inhibitors of nuclear export (SINEs) are a class of compounds that function by inhibiting the nuclear export protein exportin 1 (XPO1). The XPO1 protein is an important regulator of key inflammatory and neurological factors that drive inflammation and neurotoxicity in various neurological and neuromuscular diseases. Here, we demonstrate that SINE compound KPT-350 can ameliorate dystrophic-associated pathologies in the muscles of DMD models of zebrafish and mice. Thus, SINE compounds are a promising novel strategy for blocking dystrophic symptoms and could be used in combinatorial treatments for DMD.
Collapse
Affiliation(s)
- Rylie M Hightower
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA; UAB Center for Exercise Medicine (UCEM), Birmingham, AL 35294, USA
| | - Andrea L Reid
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
| | - Devin E Gibbs
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, MA 02115, USA
| | - Yimin Wang
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA
| | - Jeffrey J Widrick
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, MA 02115, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics at Boston Children's Hospital, Boston, MA 02115, USA; Department of Genetics at Harvard Medical School, Boston, MA 02115, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; The Manton Center for Orphan Disease Research at Boston Children's Hospital, Boston, MA 02115, USA
| | - Jenna M Kastenschmidt
- Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California-Irvine, Irvine, CA 92697, USA
| | - S Armando Villalta
- Department of Physiology and Biophysics, University of California-Irvine, Irvine, CA 92697, USA; Institute for Immunology, University of California-Irvine, Irvine, CA 92697, USA
| | - Thomas van Groen
- Department of Cell, Developmental, and Integrative Biology, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Hua Chang
- Karyopharm Therapeutics, Newton, MA 02459, USA
| | | | | | | | - Matthew S Alexander
- Department of Pediatrics, Division of Neurology, University of Alabama at Birmingham and Children's of Alabama, Birmingham, AL 35294, USA; UAB Center for Exercise Medicine (UCEM), Birmingham, AL 35294, USA; Department of Genetics at the University of Alabama at Birmingham, Birmingham, AL 35294, USA; Civitan International Research Center at the University of Alabama at Birmingham, Birmingham, AL 35294, USA.
| |
Collapse
|
27
|
Cultured hippocampal neurons of dystrophic mdx mice respond differently from those of wild type mice to an acute treatment with corticosterone. Exp Cell Res 2020; 386:111715. [PMID: 31711918 DOI: 10.1016/j.yexcr.2019.111715] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 11/04/2019] [Accepted: 11/05/2019] [Indexed: 12/27/2022]
Abstract
Duchenne muscular dystrophy is a lethal genetic disease characterised by progressive degeneration of skeletal muscles induced by deficiency of dystrophin, a cytoskeletal protein expressed in myocytes and in certain neuron populations. The severity of the neurological disorder varies in humans and animal models owing to dysfunction in numerous brain areas, including the hippocampus. Cyclic treatments with high-dose glucocorticoids remain a major pharmacological approach for treating the disease; however, elevated systemic levels of either stress-induced or exogenously administered anti-inflammatory molecules dramatically affect hippocampal activity. In this study, we analysed and compared the response of hippocampal neurons isolated from wild-type and dystrophic mdx mice to acute administration of corticosterone in vitro, without the influence of other glucocorticoid-regulated processes. Our results showed that in neurons of mdx mice, both the genomic and intracellular signalling-mediated responses to corticosterone were affected compared to those in wild-type animals, evoking the characteristic response to detrimental chronic glucocorticoid exposure. Responsiveness to glucocorticoids is, therefore, another function of hippocampal neurons possibly affected by deficiency of Dp427 since embryonic development. Knowing the pivotal role of hippocampus in stress hormone signalling, attention should be paid to the effects that prolonged glucocorticoid treatments may have on this and other brain areas of DMD patients.
Collapse
|
28
|
Finkler JM, Carvalho SC, Santo Neto H, Marques MJ. Cardiac and skeletal muscle changes associated with rosuvastatin therapy in dystrophic
mdx
mice. Anat Rec (Hoboken) 2019; 303:2202-2212. [DOI: 10.1002/ar.24341] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Revised: 11/08/2019] [Accepted: 11/14/2019] [Indexed: 12/31/2022]
Affiliation(s)
- Júlia M.G. Finkler
- Department of Structural and Functional Biology Institute of Biology, University of Campinas (UNICAMP) Campinas Brazil
| | - Samara C. Carvalho
- Department of Structural and Functional Biology Institute of Biology, University of Campinas (UNICAMP) Campinas Brazil
| | - Humberto Santo Neto
- Department of Structural and Functional Biology Institute of Biology, University of Campinas (UNICAMP) Campinas Brazil
| | - Maria J. Marques
- Department of Structural and Functional Biology Institute of Biology, University of Campinas (UNICAMP) Campinas Brazil
| |
Collapse
|
29
|
Sun J, Ma X, Chu HT, Feng B, Tuan RS, Jiang Y. Biomaterials and Advanced Biofabrication Techniques in hiPSCs Based Neuromyopathic Disease Modeling. Front Bioeng Biotechnol 2019; 7:373. [PMID: 31850331 PMCID: PMC6895005 DOI: 10.3389/fbioe.2019.00373] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 11/13/2019] [Indexed: 12/22/2022] Open
Abstract
Induced pluripotent stem cells (iPSCs) are reprogrammed somatic cells by defined factors, and have great application potentials in tissue regeneration and disease modeling. Biomaterials have been widely used in stem cell-based studies, and are involved in human iPSCs based studies, but they were not enough emphasized and recognized. Biomaterials can mimic the extracellular matrix and microenvironment, and act as powerful tools to promote iPSCs proliferation, differentiation, maturation, and migration. Many classic and advanced biofabrication technologies, such as cell-sheet approach, electrospinning, and 3D-bioprinting, are used to provide physical cues in macro-/micro-patterning, and in combination with other biological factors to support iPSCs applications. In this review, we highlight the biomaterials and fabrication technologies used in human iPSC-based tissue engineering to model neuromyopathic diseases, particularly those with genetic mutations, such as Duchenne Muscular Dystrophy (DMD), Congenital Heart Diseases (CHD) and Alzheimer's disease (AD).
Collapse
Affiliation(s)
- Jing Sun
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Xun Ma
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ho Ting Chu
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Bo Feng
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China.,Key Laboratory for Regenerative Medicine, Ministry of Education, Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Rocky S Tuan
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yangzi Jiang
- Faculty of Medicine, School of Biomedical Sciences, Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
30
|
Eccentric exercise results in a prolonged increase in interleukin-6 and tumor necrosis factor-α levels in rat skeletal muscle. J Muscle Res Cell Motil 2019; 40:379-387. [PMID: 31520264 DOI: 10.1007/s10974-019-09554-6] [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: 08/24/2018] [Accepted: 09/06/2019] [Indexed: 12/20/2022]
Abstract
Interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-α) are well-known cytokines with pro-inflammatory capabilities, and have been shown to be involved in adaptation to exercise as multifaceted myokines. However, the precise role of IL-6 and TNF-α during exercise-induced skeletal muscle injury and subsequent repair processes is not fully understood. In this study, IL-6 and TNF-α were examined in soleus muscles at the gene and protein levels using in situ hybridization and immunohistochemical staining, respectively, and serum levels of IL-6 and TNF-α were determined before and after a 90-min downhill running session in rats. There were no changes in serum levels of IL-6 and TNF-α after exercise, but IL-6 and TNF-α mRNA increased and maintained high expression in muscles for 1-2 weeks after exercise. IL-6 and TNF-a mRNAs were identified in both the cytoplasm and the nuclei of myocytes, as well as in invading inflammatory cells. IL-6 and TNF-α protein mainly distributed in cytoplasm unevenly and had a prolonged expression until 2 weeks after eccentric exercise. Our results demonstrate that there is increased IL-6 and TNF-α expression in skeletal muscle that is induced by eccentric exercise and that the high expression of IL-6 and TNF-α in the long-term phase after eccentric exercise may be more involved in the subsequent recovery of damaged muscle.
Collapse
|
31
|
Ouisse LH, Remy S, Lafoux A, Larcher T, Tesson L, Chenouard V, Guillonneau C, Brusselle L, Vimond N, Rouger K, Péréon Y, Chenouard A, Gras-Le Guen C, Braudeau C, Josien R, Huchet C, Anegon I. Immunophenotype of a Rat Model of Duchenne's Disease and Demonstration of Improved Muscle Strength After Anti-CD45RC Antibody Treatment. Front Immunol 2019; 10:2131. [PMID: 31552055 PMCID: PMC6746111 DOI: 10.3389/fimmu.2019.02131] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 08/23/2019] [Indexed: 01/25/2023] Open
Abstract
Corticosteroids (CS) are standard therapy for the treatment of Duchenne's muscular dystrophy (DMD). Even though they decrease inflammation, they have limited efficacy and are associated with significant side effects. There is therefore the need for new protolerogenic treatments to replace CS. Dystrophin-deficient rats (Dmdmdx ) closely resemble the pathological phenotype of DMD patients. We performed the first Immunophenotyping of Dmdmdx rats and showed leukocyte infiltration in skeletal and cardiac muscles, which consisted mostly of macrophages and T cells including CD45RChigh T cells. Muscles of DMD patients also contain elevated CD45RChigh T cells. We treated Dmdmdx rats with an anti-CD45RC MAb used in previous studies to deplete CD45RChigh T cells and induce immune tolerance in models of organ transplantation. Treatment of young Dmdmdx rats with anti-CD45RC MAb corrected skeletal muscle strength and was associated with depletion of CD45RChigh T cells with no side effects. Treatment of young Dmdmdx rats with prednisolone resulted in increase in skeletal muscle strength but also severe growth retardation. In conclusion, anti-CD45RC MAb treatment has potential in the treatment of DMD and might eventually result in reduction or elimination of CS use.
Collapse
Affiliation(s)
- Laure-Hélène Ouisse
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
- Transgenesis Rat ImmunoPhenomic Facility, CRTI UMR 1064, Nantes, France
| | - Séverine Remy
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
- Transgenesis Rat ImmunoPhenomic Facility, CRTI UMR 1064, Nantes, France
| | - Aude Lafoux
- THERASSAY CAPACITES, Université de Nantes, Nantes, France
| | - Thibaut Larcher
- INRA, UMR703 APEX, Oniris, Ecole Nationale Vétérinaire, Agro-alimentaire et de l'alimentation, Nantes, France
| | - Laurent Tesson
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
- Transgenesis Rat ImmunoPhenomic Facility, CRTI UMR 1064, Nantes, France
| | - Vanessa Chenouard
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
- Transgenesis Rat ImmunoPhenomic Facility, CRTI UMR 1064, Nantes, France
| | - Carole Guillonneau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Lucas Brusselle
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
- Transgenesis Rat ImmunoPhenomic Facility, CRTI UMR 1064, Nantes, France
| | - Nadège Vimond
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
| | - Karl Rouger
- INRA, UMR703 APEX, Oniris, Ecole Nationale Vétérinaire, Agro-alimentaire et de l'alimentation, Nantes, France
| | - Yann Péréon
- Reference Centre for Neuromuscular Diseases AOC, CHU Nantes, Nantes, France
| | - Alexis Chenouard
- Pediatric Intensive Care, Hôpital Mère Enfant, CHU Nantes, Nantes, France
| | | | - Cécile Braudeau
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
- CIMNA, Laboratoire d'Immunologie, CHU Nantes, Nantes, France
| | - Régis Josien
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
- CIMNA, Laboratoire d'Immunologie, CHU Nantes, Nantes, France
| | - Corinne Huchet
- THERASSAY CAPACITES, Université de Nantes, Nantes, France
- Thérapie Génique Translationnelle des Maladies Génétiques, INSERM UMR 1089, Nantes, France
| | - Ignacio Anegon
- Centre de Recherche en Transplantation et Immunologie UMR 1064, INSERM, Université de Nantes, Nantes, France
- Institut de Transplantation Urologie Néphrologie (ITUN), CHU Nantes, Nantes, France
- Transgenesis Rat ImmunoPhenomic Facility, CRTI UMR 1064, Nantes, France
| |
Collapse
|
32
|
Oishi Y, Manabe I. Macrophages in inflammation, repair and regeneration. Int Immunol 2019; 30:511-528. [PMID: 30165385 DOI: 10.1093/intimm/dxy054] [Citation(s) in RCA: 401] [Impact Index Per Article: 66.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 08/22/2018] [Indexed: 12/12/2022] Open
Abstract
Tissue injury triggers a complex series of cellular responses, starting from inflammation activated by tissue and cell damage and proceeding to healing. By clearing cell debris, activating and resolving inflammation and promoting fibrosis, macrophages play key roles in most, if not all, phases of the response to injury. Recent studies of the mechanisms underlying the initial inflammation and later tissue regeneration and repair revealed that macrophages bridge these processes in part by supporting and activating stem/progenitor cells, clearing damaged tissue, remodeling extracellular matrix to prepare scaffolding for regeneration and promoting angiogenesis. However, macrophages also have a central role in the development of pathology induced by failed resolution (e.g. chronic inflammation) and excessive scarring. In this review, we summarize the activities of macrophages in inflammation and healing in response to acute injury in tissues with differing regenerative capacities. While macrophages lead similar processes in response to tissue injury in these tissues, their priorities and the consequences of their activities differ among tissues. Moreover, the magnitude, nature and duration of injury also greatly affect cellular responses and healing processes. In particular, continuous injury and/or failed resolution of inflammation leads to chronic ailments in which macrophage activities may become detrimental.
Collapse
Affiliation(s)
- Yumiko Oishi
- Department of Biochemistry & Molecular Biology, Nippon Medical School, Bunkyo-ku, Tokyo, Japan
| | - Ichiro Manabe
- Department of Disease Biology and Molecular Medicine, Chiba University Graduate School of Medicine, Chuo-ku, Chiba, Japan
| |
Collapse
|
33
|
Kranig SA, Tschada R, Braun M, Patry C, Pöschl J, Frommhold D, Hudalla H. Dystrophin deficiency promotes leukocyte recruitment in mdx mice. Pediatr Res 2019; 86:188-194. [PMID: 31091530 DOI: 10.1038/s41390-019-0427-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 04/11/2019] [Accepted: 05/03/2019] [Indexed: 01/08/2023]
Abstract
BACKGROUND A growing body of evidence defines inflammation as a hallmark feature of disease pathogenesis of Duchenne muscular dystrophy. To tailor potential immune modulatory interventions, a better understanding of immune dysregulation in Duchenne muscular dystrophy is needed. We now asked whether dystrophin deficiency affects the cascade of leukocyte recruitment. METHODS We performed intravital microscopy on the cremaster muscle of wild-type and dystrophin-deficient mdx mice. Recruitment was triggered by preparation alone (traumatic inflammation) or in combination with scrotal TNFα injections. Neutrophilic infiltration of the cremaster muscle was assessed on tissue sections. Integrin expression on circulating neutrophils and serum levels of pro-inflammatory cytokines were measured by flow cytometry. RESULTS Mdx mice show increased rolling and adhesion at baseline (traumatic inflammation) and a more profound response upon TNFα injection compared with wild-type animals. In both models, neutrophilic infiltration of the cremaster muscle is increased. Upregulation of the integrins LFA-1 and Mac-1 on circulating leukocytes and pro-inflammatory cytokines IL-6 and CCL2 in the serum points toward systemically altered immune regulation in mdx mice. CONCLUSION We are the first to show exaggerated activation of the leukocyte recruitment cascade in a dystrophin-deficient organism in vivo.
Collapse
Affiliation(s)
- Simon Alexander Kranig
- Department of Neonatology, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany
| | - Raphaela Tschada
- Department of Neonatology, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany
| | - Maylis Braun
- Department of Neonatology, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany
| | - Christian Patry
- Department of General Pediatrics, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany
| | - Johannes Pöschl
- Department of Neonatology, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany
| | - David Frommhold
- Klinik für Kinderheilkunde und Jugendmedizin, 87700, Memmingen, Germany
| | - Hannes Hudalla
- Department of Neonatology, Heidelberg University Children's Hospital, 69120, Heidelberg, Germany.
| |
Collapse
|
34
|
Haupenthal DPDS, Possato JC, Zaccaron RP, Mendes C, Rodrigues MS, Nesi RT, Pinho RA, Feuser PE, Machado-de-Ávila RA, Comim CM, Silveira PCL. Effects of chronic treatment with gold nanoparticles on inflammatory responses and oxidative stress in Mdx mice. J Drug Target 2019; 28:46-54. [PMID: 31046473 DOI: 10.1080/1061186x.2019.1613408] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive hereditary myopathy characterised by progressive muscle degeneration in male children. As a consequence of DMD, increased inflammation and oxidative stress occur in muscle tissue along with morphological changes. Several studies have reported anti-inflammatory and antioxidant effects of gold nanoparticles (GNP) in muscle injury models. The objective of this study was to evaluate these effects along with the impacts of the disease on histopathological changes following chronic administration of GNP to Mdx mice. Two-month-old Mdx mice were separated into five groups of eight individuals each, as follows: wild-type (WT), Mdx-modified without treatment, Mdx + 2.5 mg/kg GNP, Mdx + 7.0 mg/kg GNP and Mdx + 21 mg/kg GNP. GNP with a mean diameter of 20 nm were injected subcutaneously at concentrations of 2.5, 7.0 and 21 mg/kg. Treatments continued for 30 d with injections administered at 48-h intervals. Twenty-four hours after the last injection, the animals were killed and the central region of the gastrocnemius muscle was surgically removed. Chronic administration of GNP reduced inflammation in the gastrocnemius muscle of Mdx mice and reduced morphological alterations due to inflammatory responses to muscular dystrophy. In addition, GNP also demonstrated antioxidant potential by reducing the production of reactive oxygen and nitrogen species, reducing oxidative damage and improving antioxidant activity.
Collapse
Affiliation(s)
| | - Jonathann Corrêa Possato
- Laboratory of Experimental Phisiopatology, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Rubya Pereira Zaccaron
- Laboratory of Experimental Phisiopatology, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | - Carolini Mendes
- Laboratory of Experimental Phisiopatology, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | | | - Renata Tiscoski Nesi
- Laboratory of Exercise Biochemistry in Health, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Ricardo Aurino Pinho
- Laboratory of Exercise Biochemistry in Health, School of Medicine, Pontifícia Universidade Católica do Paraná, Curitiba, Brazil
| | - Paulo Emilio Feuser
- Laboratory of Experimental Phisiopatology, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| | | | - Clarissa M Comim
- Research Group of Experimental Neuropathology, Laboratory of Experimental Neuroscience, University of South Santa Catarina, Palhoça, Brazil
| | - Paulo Cesar Lock Silveira
- Laboratory of Experimental Phisiopatology, Universidade do Extremo Sul Catarinense, Criciúma, Brazil
| |
Collapse
|
35
|
Hughes MC, Ramos SV, Turnbull PC, Rebalka IA, Cao A, Monaco CM, Varah NE, Edgett BA, Huber JS, Tadi P, Delfinis LJ, Schlattner U, Simpson JA, Hawke TJ, Perry CG. Early myopathy in Duchenne muscular dystrophy is associated with elevated mitochondrial H 2 O 2 emission during impaired oxidative phosphorylation. J Cachexia Sarcopenia Muscle 2019; 10:643-661. [PMID: 30938481 PMCID: PMC6596403 DOI: 10.1002/jcsm.12405] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 12/13/2018] [Accepted: 01/09/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Muscle wasting and weakness in Duchenne muscular dystrophy (DMD) causes severe locomotor limitations and early death due in part to respiratory muscle failure. Given that current clinical practice focuses on treating secondary complications in this genetic disease, there is a clear need to identify additional contributions in the aetiology of this myopathy for knowledge-guided therapy development. Here, we address the unresolved question of whether the complex impairments observed in DMD are linked to elevated mitochondrial H2 O2 emission in conjunction with impaired oxidative phosphorylation. This study performed a systematic evaluation of the nature and degree of mitochondrial-derived H2 O2 emission and mitochondrial oxidative dysfunction in a mouse model of DMD by designing in vitro bioenergetic assessments that attempt to mimic in vivo conditions known to be critical for the regulation of mitochondrial bioenergetics. METHODS Mitochondrial bioenergetics were compared with functional and histopathological indices of myopathy early in DMD (4 weeks) in D2.B10-DMDmdx /2J mice (D2.mdx)-a model that demonstrates severe muscle weakness. Adenosine diphosphate's (ADP's) central effect of attenuating H2 O2 emission while stimulating respiration was compared under two models of mitochondrial-cytoplasmic phosphate exchange (creatine independent and dependent) in muscles that stained positive for membrane damage (diaphragm, quadriceps, and white gastrocnemius). RESULTS Pathway-specific analyses revealed that Complex I-supported maximal H2 O2 emission was elevated concurrent with a reduced ability of ADP to attenuate emission during respiration in all three muscles (mH2 O2 : +17 to +197% in D2.mdx vs. wild type). This was associated with an impaired ability of ADP to stimulate respiration at sub-maximal and maximal kinetics (-17 to -72% in D2.mdx vs. wild type), as well as a loss of creatine-dependent mitochondrial phosphate shuttling in diaphragm and quadriceps. These changes largely occurred independent of mitochondrial density or abundance of respiratory chain complexes, except for quadriceps. This muscle was also the only one exhibiting decreased calcium retention capacity, which indicates increased sensitivity to calcium-induced permeability transition pore opening. Increased H2 O2 emission was accompanied by a compensatory increase in total glutathione, while oxidative stress markers were unchanged. Mitochondrial bioenergetic dysfunctions were associated with induction of mitochondrial-linked caspase 9, necrosis, and markers of atrophy in some muscles as well as reduced hindlimb torque and reduced respiratory muscle function. CONCLUSIONS These results provide evidence that Complex I dysfunction and loss of central respiratory control by ADP and creatine cause elevated oxidant generation during impaired oxidative phosphorylation. These dysfunctions may contribute to early stage disease pathophysiology and support the growing notion that mitochondria are a potential therapeutic target in this disease.
Collapse
Affiliation(s)
- Meghan C. Hughes
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| | - Sofhia V. Ramos
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| | - Patrick C. Turnbull
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| | - Irena A. Rebalka
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonONCanada
| | - Andrew Cao
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonONCanada
| | - Cynthia M.F. Monaco
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonONCanada
| | - Nina E. Varah
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonONCanada
| | - Brittany A. Edgett
- Department of Human Health and Nutritional Sciences and Cardiovascular Research GroupUniversity of GuelphGuelphONCanada
| | - Jason S. Huber
- Department of Human Health and Nutritional Sciences and Cardiovascular Research GroupUniversity of GuelphGuelphONCanada
| | - Peyman Tadi
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| | - Luca J. Delfinis
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| | - U. Schlattner
- Laboratory of Fundamental and Applied Bioenergetics (LBFA) and SFR Environmental and Systems Biology (BEeSy)University Grenoble AlpesGrenobleFrance
| | - Jeremy A. Simpson
- Department of Human Health and Nutritional Sciences and Cardiovascular Research GroupUniversity of GuelphGuelphONCanada
| | - Thomas J. Hawke
- Department of Pathology and Molecular MedicineMcMaster UniversityHamiltonONCanada
| | - Christopher G.R. Perry
- School of Kinesiology and Health Science, Muscle Health Research Centre, 344 Norman Bethune CollegeYork UniversityTorontoONCanada
| |
Collapse
|
36
|
Marinkovic M, Fuoco C, Sacco F, Cerquone Perpetuini A, Giuliani G, Micarelli E, Pavlidou T, Petrilli LL, Reggio A, Riccio F, Spada F, Vumbaca S, Zuccotti A, Castagnoli L, Mann M, Gargioli C, Cesareni G. Fibro-adipogenic progenitors of dystrophic mice are insensitive to NOTCH regulation of adipogenesis. Life Sci Alliance 2019; 2:e201900437. [PMID: 31239312 PMCID: PMC6599969 DOI: 10.26508/lsa.201900437] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 06/07/2019] [Indexed: 12/14/2022] Open
Abstract
Fibro-adipogenic progenitors (FAPs) promote satellite cell differentiation in adult skeletal muscle regeneration. However, in pathological conditions, FAPs are responsible for fibrosis and fatty infiltrations. Here we show that the NOTCH pathway negatively modulates FAP differentiation both in vitro and in vivo. However, FAPs isolated from young dystrophin-deficient mdx mice are insensitive to this control mechanism. An unbiased mass spectrometry-based proteomic analysis of FAPs from muscles of wild-type and mdx mice suggested that the synergistic cooperation between NOTCH and inflammatory signals controls FAP differentiation. Remarkably, we demonstrated that factors released by hematopoietic cells restore the sensitivity to NOTCH adipogenic inhibition in mdx FAPs. These results offer a basis for rationalizing pathological ectopic fat infiltrations in skeletal muscle and may suggest new therapeutic strategies to mitigate the detrimental effects of fat depositions in muscles of dystrophic patients.
Collapse
Affiliation(s)
| | - Claudia Fuoco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | | | - Giulio Giuliani
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Elisa Micarelli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Alessio Reggio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Federica Riccio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Filomena Spada
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Simone Vumbaca
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Matthias Mann
- Department of Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Cesare Gargioli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Fondazione Santa Lucia, Rome, Italy
| |
Collapse
|
37
|
Comim CM, Ventura L, Freiberger V, Dias P, Bragagnolo D, Dutra ML, Amaral RA, Camargo-Fagundes ALS, Reis PA, Castro-Faria-Neto HC, Vainzof M, Rosa MI. Neurocognitive Impairment in mdx Mice. Mol Neurobiol 2019; 56:7608-7616. [PMID: 31077034 DOI: 10.1007/s12035-019-1573-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 03/20/2019] [Indexed: 12/16/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a neuromuscular disorder that affects muscles and also the brain, resulting in memory and behavioral problems. In the pathogenesis of DMD, inflammation is an important factor during the degenerative process. However, the involvement of the brain is still unclear. Therefore, the objective of this study is to evaluate the cognitive involvement, BDNF levels, cytokine levels through the levels of TNF-α and IL-1β, the myeloperoxidase (MPO) activity, and the expression of proteins postsynaptic density (PSD)-95 and synaptophysin in the brain of mdx mice. To this aim, we used adult mdx mice. It was observed that mdx mice presented deficits on the habituation, aversive, and object recognition memory. These animals also had a depression-like behavior and an anxiety-like behavior, a decrease of BDNF levels, an increase in the levels of TNF-α and IL-1β, an increase of MPO activity, and an overexpression of synaptophysin and PSD-95 in brain tissue. In conclusion, these data show that mdx mice possibly present a neuroinflammatory component and the involvement of synaptic proteins associated to memory storage and restoring process impairment as well as a depressive- and anxiety-like behavior.
Collapse
Affiliation(s)
- Clarissa M Comim
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, SC, Brazil.
| | - Letícia Ventura
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, SC, Brazil
| | - Viviane Freiberger
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, SC, Brazil
| | - Paula Dias
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, SC, Brazil
| | - Daiane Bragagnolo
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, SC, Brazil
| | - Matheus L Dutra
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, SC, Brazil
| | - Ricardo A Amaral
- Research Group on Neurodevelopment of Childhood and Adolescence, Laboratory of Experimental Neuroscience, Postgraduate Program in Health Sciences, University of South Santa Catarina, Palhoça, SC, Brazil
| | - Ana Lucia S Camargo-Fagundes
- Laboratory of Epidemiology, Postgraduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil
| | - Patrícia A Reis
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz/IOC/FIOCRUZ-Manguinhos, Rio de Janeiro, Brazil
| | - Hugo C Castro-Faria-Neto
- Laboratory of Immunopharmacology, Instituto Oswaldo Cruz/IOC/FIOCRUZ-Manguinhos, Rio de Janeiro, Brazil
| | - Mariz Vainzof
- Human Genome Research Center, Biosciences Institute, University of São Paulo, São Paulo, SP, Brazil
| | - Maria I Rosa
- Laboratory of Epidemiology, Postgraduate Program in Health Sciences, Health Sciences Unit, University of Southern Santa Catarina, Criciúma, SC, 88806-000, Brazil
| |
Collapse
|
38
|
Hoxha M. Duchenne muscular dystrophy: Focus on arachidonic acid metabolites. Biomed Pharmacother 2019; 110:796-802. [DOI: 10.1016/j.biopha.2018.12.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 12/07/2018] [Accepted: 12/07/2018] [Indexed: 12/11/2022] Open
|
39
|
Hermes TDA, Kido LA, Macedo AB, Mizobuti DS, Moraes LHR, Somazz MC, Cagnon VHA, Minatel E. Sex influences diaphragm muscle response in exercised mdx mice. Cell Biol Int 2018; 42:1611-1621. [PMID: 30238549 DOI: 10.1002/cbin.11057] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 09/16/2018] [Indexed: 12/26/2022]
Abstract
Physical exercise promotes increased muscle damage in the mdx mice, the experimental model of Duchenne muscular dystrophy. Studies suggest that the estrogen level in females makes them less susceptible to muscle injuries. The aim of this study was to characterize the diaphragm (DIA) muscle response to physical exercise in male and female mdx mice. The animals were divided into four groups: female sedentary mdx; male sedentary mdx; female mdx submitted to exercise; and male mdx mice submitted to exercise. Blood samples were used to determine creatine kinase (CK). Regenerated muscle fibers were indicated by the presence of central nucleus and also inflammation areas were determined in DIA muscle sections. The alpha and beta estrogen receptors (ER) were determined by means of immunohistochemistry evaluation in the dystrophic DIA muscle. Male mdx animals submitted to exercise showed increased CK levels and inflammatory area. The quantification of regenerated fibers was higher in male animals, submitted or not to physical exercise. Greater alpha and beta ER expression was verified in the females submitted to exercise in the DIA muscle than in the other experimental groups. Therefore, estrogen may have contributed to the prevention of increased inflammatory process and DIA injury in females submitted to exercise.
Collapse
Affiliation(s)
- Túlio de Almeida Hermes
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil.,Centro Regional Universitário de Espírito Santo do Pinhal (UNIPINHAL), Espírito Santo do Pinhal, São Paulo 13990-000, Brazil
| | - Larissa Akemi Kido
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | - Aline Barbosa Macedo
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | - Daniela Sayuri Mizobuti
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | - Luiz Henrique Rapucci Moraes
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | - Marco Cesar Somazz
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil.,Centro Regional Universitário de Espírito Santo do Pinhal (UNIPINHAL), Espírito Santo do Pinhal, São Paulo 13990-000, Brazil
| | - Valéria Helena Alves Cagnon
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology, Institute of Biology, State University of Campinas (UNICAMP), Campinas, São Paulo 13083-970, Brazil
| |
Collapse
|
40
|
Juhas M, Abutaleb N, Wang JT, Ye J, Shaikh Z, Sriworarat C, Qian Y, Bursac N. Incorporation of macrophages into engineered skeletal muscle enables enhanced muscle regeneration. Nat Biomed Eng 2018; 2:942-954. [PMID: 30581652 DOI: 10.1038/s41551-018-0290-2] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adult skeletal muscle has a robust capacity for self-repair, owing to synergies between muscle satellite cells and the immune system. In vitro models of muscle self-repair would facilitate the basic understanding of muscle regeneration and the screening of therapies for muscle disease. Here, we show that the incorporation of macrophages into muscle tissues engineered from adult-rat myogenic cells enables near-complete structural and functional repair after cardiotoxic injury in vitro. First, we show that-in contrast with injured neonatal-derived engineered muscle-adult-derived engineered muscle fails to properly self-repair after injury, even when treated with pro-regenerative cytokines. We then show that rat bone-marrow-derived macrophages or human blood-derived macrophages resident within the in vitro engineered tissues stimulate muscle satellite cell-mediated myogenesis while significantly limiting myofibre apoptosis and degeneration. Moreover, bone-marrow-derived macrophages within engineered tissues implanted in a mouse dorsal window-chamber model augmented blood vessel ingrowth, cell survival, muscle regeneration and contractile function.
Collapse
Affiliation(s)
- Mark Juhas
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nadia Abutaleb
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jason T Wang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Jean Ye
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Zohaib Shaikh
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | | | - Ying Qian
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA. .,Regeneration Next, Duke University, Durham, NC, USA.
| |
Collapse
|
41
|
Abstract
The immune response to acute muscle damage is important for normal repair. However, in chronic diseases such as many muscular dystrophies, the immune response can amplify pathology and play a major role in determining disease severity. Muscular dystrophies are inheritable diseases that vary tremendously in severity, but share the progressive loss of muscle mass and function that can be debilitating and lethal. Mutations in diverse genes cause muscular dystrophy, including genes that encode proteins that maintain membrane strength, participate in membrane repair, or are components of the extracellular matrix or the nuclear envelope. In this article, we explore the hypothesis that an important feature of many muscular dystrophies is an immune response adapted to acute, infrequent muscle damage that is misapplied in the context of chronic injury. We discuss the involvement of the immune system in the most common muscular dystrophy, Duchenne muscular dystrophy, and show that the immune system influences muscle death and fibrosis as disease progresses. We then present information on immune cell function in other muscular dystrophies and show that for many muscular dystrophies, release of cytosolic proteins into the extracellular space may provide an initial signal, leading to an immune response that is typically dominated by macrophages, neutrophils, helper T-lymphocytes, and cytotoxic T-lymphocytes. Although those features are similar in many muscular dystrophies, each muscular dystrophy shows distinguishing features in the magnitude and type of inflammatory response. These differences indicate that there are disease-specific immunomodulatory molecules that determine response to muscle cell damage caused by diverse genetic mutations. © 2018 American Physiological Society. Compr Physiol 8:1313-1356, 2018.
Collapse
Affiliation(s)
- James G. Tidball
- Molecular, Cellular & Integrative Physiology Program, University of California, Los Angeles, California, USA
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, University of California, Los Angeles, California, USA
| | - Steven S. Welc
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| | - Michelle Wehling-Henricks
- Department of Integrative Biology and Physiology, University of California, Los Angeles, California, USA
| |
Collapse
|
42
|
Morgan JE, Prola A, Mariot V, Pini V, Meng J, Hourde C, Dumonceaux J, Conti F, Relaix F, Authier FJ, Tiret L, Muntoni F, Bencze M. Necroptosis mediates myofibre death in dystrophin-deficient mice. Nat Commun 2018; 9:3655. [PMID: 30194302 PMCID: PMC6128848 DOI: 10.1038/s41467-018-06057-9] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Accepted: 08/10/2018] [Indexed: 12/25/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a severe degenerative disorder caused by mutations in the dystrophin gene. Dystrophin-deficient muscles are characterised by progressive myofibre necrosis in which inflammation plays a deleterious role. However, the molecular mechanisms underlying inflammation-induced necrosis in muscle cells are unknown. Here we show that necroptosis is a mechanism underlying myofibre death in dystrophin-deficient muscle. RIPK1, RIPK3 and MLKL are upregulated in dystrophic mouse myofibres. In human DMD samples, there is strong immunoreactivity to RIPK3 and phospho-MLKL in myofibres. In vitro, TNFα can elicit necroptosis in C2C12 myoblasts, and RIPK3 overexpression sensitises myoblasts to undergo TNF-induced death. Furthermore, genetic ablation of Ripk3 in mdx mice reduces myofibre degeneration, inflammatory infiltrate, and muscle fibrosis, and eventually improves muscle function. These findings provide the first evidence of necroptotic cell death in a disease affecting skeletal muscle and identify RIPK3 as a key player in the degenerative process in dystrophin-deficient muscles.
Collapse
Affiliation(s)
- Jennifer E Morgan
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK.
| | - Alexandre Prola
- U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, ENVA, EFS, Créteil, 94000, France
| | - Virginie Mariot
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, 30 Guilford Street, London, WC1N 1EH, UK
| | - Veronica Pini
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Jinhong Meng
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Christophe Hourde
- Inter-University Laboratory of Human Movement Biology (LIBM)-EA7424, Université Savoie Mont Blanc, Campus Scientifique Technolac, 73376, Le Bourget du Lac Cedex, France
| | - Julie Dumonceaux
- NIHR Biomedical Research Centre, University College London, Great Ormond Street Institute of Child Health and Great Ormond Street Hospital NHS Trust, 30 Guilford Street, London, WC1N 1EH, UK
| | - Francesco Conti
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Frederic Relaix
- U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, ENVA, EFS, Créteil, 94000, France.,Nord/Est/Ile-de-France Reference Centre for Neuromuscular Diseases, Henri Mondor University Hospital (APHP), 94000, Créteil, France
| | - Francois-Jerôme Authier
- U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, ENVA, EFS, Créteil, 94000, France.,Nord/Est/Ile-de-France Reference Centre for Neuromuscular Diseases, Henri Mondor University Hospital (APHP), 94000, Créteil, France
| | - Laurent Tiret
- U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, ENVA, EFS, Créteil, 94000, France
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK
| | - Maximilien Bencze
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, London, WC1N 1EH, UK. .,U955-IMRB, Team 10, Biology of the Neuromuscular System, Inserm, UPEC, ENVA, EFS, Créteil, 94000, France.
| |
Collapse
|
43
|
Srikuea R, Suhatcho K. Impact of intramuscular administration of lipid-soluble and water-soluble vehicles into regenerating muscle at the distinct phases of skeletal muscle regeneration. J Physiol Sci 2018; 68:647-661. [PMID: 29134575 PMCID: PMC10717534 DOI: 10.1007/s12576-017-0576-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 10/28/2017] [Indexed: 12/30/2022]
Abstract
Interpretation on the effectiveness of potential substances to enhance skeletal muscle regeneration is difficult if an inappropriate vehicle is administered, since vehicle administration can directly enhance or suppress regenerative capacity. In the current study, intramuscular administration of lipid-soluble and water-soluble vehicles into regenerating muscle at the distinct phases of skeletal muscle regeneration (regenerative vs. remodeling) were investigated. Tested vehicles included lipid-soluble [olive oil, (0.1, 1, 5, and 40%) dimethyl sulfoxide (DMSO), and 40% propylene glycol (PG)] and water-soluble [0.9% NaCl, PBS, 0.1% ethanol, and distilled water]. Skeletal muscle regeneration was induced by 1.2% BaCl2 injection to the tibialis anterior muscle of 10-week-old C57BL/6 male mice. Histological features, skeletal muscle stem cell activity, regenerating muscle fiber formation, angiogenesis, extracellular matrix remodeling, and macrophage infiltration were examined. The results revealed repeated administration of 40% DMSO and 40% PG causes significant recurrent muscle injury, which is pronounced during the remodeling phase compared to the regenerative phase. These findings were supported by (1) massive infiltration of F4/80+ macrophages; (2) significant increase of skeletal muscle stem cell re-activation and nascent regenerating muscle fiber formation; (3) excess fibrous formation; and (4) decreased regenerating muscle fiber cross-sectional area. These deleterious effects were comparable to 2% trypsin (degenerative substance) administration and less pronounced with a single administration. Nevertheless, recurrent muscle injury was still presented with 5% DMSO administration but it can be alleviated when 0.1% DMSO was administered during the remodeling phase. In contrast, none of the tested vehicles enhanced regenerative capacity compared with IGF-1 administration. Altogether, intramuscular administration of vehicle containing high concentration of DMSO or PG could impair skeletal muscle regenerative capacity and potentially affect validation of the investigational substance.
Collapse
Affiliation(s)
- Ratchakrit Srikuea
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand.
| | - Kanokwan Suhatcho
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, 10400, Thailand
| |
Collapse
|
44
|
Al-Khalidi R, Panicucci C, Cox P, Chira N, Róg J, Young CNJ, McGeehan RE, Ambati K, Ambati J, Zabłocki K, Gazzerro E, Arkle S, Bruno C, Górecki DC. Zidovudine ameliorates pathology in the mouse model of Duchenne muscular dystrophy via P2RX7 purinoceptor antagonism. Acta Neuropathol Commun 2018; 6:27. [PMID: 29642926 PMCID: PMC5896059 DOI: 10.1186/s40478-018-0530-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 03/28/2018] [Indexed: 12/20/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is the most common inherited muscle disorder that causes severe disability and death of young men. This disease is characterized by progressive muscle degeneration aggravated by sterile inflammation and is also associated with cognitive impairment and low bone density. Given that no current treatment can improve the long-term outcome, approaches with a strong translational potential are urgently needed. Duchenne muscular dystrophy (DMD) alters P2RX7 signaling in both muscle and inflammatory cells and inhibition of this receptor resulted in a significant attenuation of muscle and non-muscle symptoms in DMDmdx mouse model. As P2RX7 is an attractive target in a range of human diseases, specific antagonists have been developed. Yet, these will require lengthy safety testing in the pediatric population of Duchenne muscular dystrophy (DMD) patients. In contrast, Nucleoside Reverse Transcriptase Inhibitors (NRTIs) can act as P2RX7 antagonists and are drugs with an established safety record, including in children. We demonstrate here that AZT (Zidovudine) inhibits P2RX7 functions acting via the same allosteric site as other antagonists. Moreover, short-term AZT treatment at the peak of disease in DMDmdx mice attenuated the phenotype without any detectable side effects. Recovery was evident in the key parameters such as reduced sarcolemma permeability confirmed by lower serum creatine kinase levels and IgG influx into myofibres, decreased inflammatory cell numbers and inflammation markers in leg and heart muscles of treated mice. Moreover, this short-term therapy had some positive impact on muscle strength in vivo and no detrimental effect on mitochondria, which is the main side-effect of Nucleoside Reverse Transcriptase Inhibitors (NRTIs). Given these results, we postulate that AZT could be quickly re-purposed for the treatment of this highly debilitating and lethal disease. This approach is not constrained by causative DMD mutations and may be effective in alleviating both muscle and non-muscle abnormalities.
Collapse
|
45
|
Ismail HM, Dorchies OM, Scapozza L. The potential and benefits of repurposing existing drugs to treat rare muscular dystrophies. Expert Opin Orphan Drugs 2018. [DOI: 10.1080/21678707.2018.1452733] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Hesham M. Ismail
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Olivier M. Dorchies
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| | - Leonardo Scapozza
- Pharmaceutical Biochemistry, School of Pharmaceutical Sciences, University of Geneva, University of Lausanne, CMU, Geneva, Switzerland
| |
Collapse
|
46
|
Collier AF, Gumerson J, Lehtimäki K, Puoliväli J, Jones JW, Kane MA, Manne S, O'Neill A, Windish HP, Ahtoniemi T, Williams BA, Albrecht DE, Bloch RJ. Effect of Ibuprofen on Skeletal Muscle of Dysferlin-Null Mice. J Pharmacol Exp Ther 2018; 364:409-419. [PMID: 29284661 PMCID: PMC5801553 DOI: 10.1124/jpet.117.244244] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 12/21/2017] [Indexed: 12/19/2022] Open
Abstract
Ibuprofen, a nonsteroidal anti-inflammatory drug, and nitric oxide (NO) donors have been reported to reduce the severity of muscular dystrophies in mice associated with the absence of dystrophin or α-sarcoglycan, but their effects on mice that are dystrophic due to the absence of dysferlin have not been examined. We have tested ibuprofen, as well as isosorbide dinitrate (ISDN), a NO donor, to learn whether used alone or together they protect dysferlin-null muscle in A/J mice from large strain injury (LSI) induced by a series of high strain lengthening contractions. Mice were maintained on chow containing ibuprofen and ISDN for 4 weeks. They were then subjected to LSI and maintained on the drugs for 3 additional days. We measured loss of torque immediately following injury and at day 3 postinjury, fiber necrosis, and macrophage infiltration at day 3 postinjury, and serum levels of the drugs at the time of euthanasia. Loss of torque immediately after injury was not altered by the drugs. However, the torque on day 3 postinjury significantly decreased as a function of ibuprofen concentration in the serum (range, 0.67-8.2 µg/ml), independent of ISDN. The effects of ISDN on torque loss at day 3 postinjury were not significant. In long-term studies of dysferlinopathic BlAJ mice, lower doses of ibuprofen had no effects on muscle morphology, but reduced treadmill running by 40%. Our results indicate that ibuprofen can have deleterious effects on dysferlin-null muscle and suggest that its use at pharmacological doses should be avoided by individuals with dysferlinopathies.
Collapse
Affiliation(s)
- Alyssa F Collier
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Jessica Gumerson
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Kimmo Lehtimäki
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Jukka Puoliväli
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Jace W Jones
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Maureen A Kane
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Sankeerth Manne
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Andrea O'Neill
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Hillarie P Windish
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Toni Ahtoniemi
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Bradley A Williams
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Douglas E Albrecht
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| | - Robert J Bloch
- Department of Physiology, University of Maryland School of Medicine, Baltimore, Maryland (A.F.C., J.G., S.M., A.O'N., R.J.B.); Charles River Laboratories, Kuopio, Finland (K.L., J.P., T.A.); Mass Spectrometry Center, Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland (M.A.K., J.W.J.); and Jain Foundation, Seattle, Washington (H.P.W., B.A.W., D.E.A.)
| |
Collapse
|
47
|
Identification of plasma interleukins as biomarkers for deflazacort and omega-3 based Duchenne muscular dystrophy therapy. Cytokine 2018; 102:55-61. [DOI: 10.1016/j.cyto.2017.12.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 12/04/2017] [Accepted: 12/05/2017] [Indexed: 01/23/2023]
|
48
|
NF-kB and Inflammatory Cytokine Signalling: Role in Skeletal Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:267-279. [PMID: 30390256 DOI: 10.1007/978-981-13-1435-3_12] [Citation(s) in RCA: 192] [Impact Index Per Article: 27.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Atrophy is a classical hallmark of an array of disorders that affect skeletal muscle, ranging from inherited dystrophies, acquired inflammatory myopathies, ageing (sarcopenia) and critical illness (sepsis). The loss of muscle mass and function in these instances is associated with disability, poor quality of life and in some cases mortality. The mechanisms which underpin muscle atrophy are complex; however, significant research has demonstrated an important role for inflammatory cytokines such as tumour necrosis factor-alpha (TNF-α), mediated by the generation of reactive oxygen species (ROS) in muscle wasting. Moreover, activation of the transcription factor nuclear factor kappa B (NF-κB) is a key lynchpin in the overall processes that mediate muscle atrophy. The significance of NF-κB as a key regulator of muscle atrophy has been emphasised by several in vivo studies, which have demonstrated that NF-κB-targeted therapies can abrogate muscle atrophy. In this chapter, we will summarise current knowledge on the role of cytokines (TNF-α) and NF-κB in the loss of muscle mass and function and highlight perspectives towards future research and potential therapies to combat muscle atrophy.
Collapse
|
49
|
Abstract
Our understanding of satellite cells, now known to be the obligate stem cells of skeletal muscle, has increased dramatically in recent years due to the introduction of new molecular, genetic, and technical resources. In addition to their role in acute repair of damaged muscle, satellite cells are of interest in the fields of aging, exercise, neuromuscular disease, and stem cell therapy, and all of these applications have driven a dramatic increase in our understanding of the activity and potential of satellite cells. However, many fundamental questions of satellite cell biology remain to be answered, including their emergence as a specific lineage, the degree and significance of heterogeneity within the satellite cell population, the roles of their interactions with other resident and infiltrating cell types during homeostasis and regeneration, and the relative roles of intrinsic vs extrinsic factors that may contribute to satellite cell dysfunction in the context of aging or disease. This review will address the current state of these open questions in satellite cell biology.
Collapse
Affiliation(s)
- Ddw Cornelison
- University of Missouri, Columbia, MO, United States; Christopher S. Bond Life Sciences Center, University of Missouri, Columbia, MO, United States.
| |
Collapse
|
50
|
Increased Circulating Levels of Interleukin-6 Induce Perturbation in Redox-Regulated Signaling Cascades in Muscle of Dystrophic Mice. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1987218. [PMID: 28845212 PMCID: PMC5563418 DOI: 10.1155/2017/1987218] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 05/31/2017] [Accepted: 06/07/2017] [Indexed: 12/26/2022]
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked genetic disease in which dystrophin gene is mutated, resulting in dysfunctional or absent dystrophin protein. The pathology of dystrophic muscle includes degeneration, necrosis with inflammatory cell invasion, regeneration, and fibrous and fatty changes. Nevertheless, the mechanisms by which the absence of dystrophin leads to muscle degeneration remain to be fully elucidated. An imbalance between oxidant and antioxidant systems has been proposed as a secondary effect of DMD. However, the significance and precise extent of the perturbation in redox signaling cascades is poorly understood. We report that mdx dystrophic mice are able to activate a compensatory antioxidant response at the presymptomatic stage of the disease. In contrast, increased circulating levels of IL-6 perturb the redox signaling cascade, even prior to the necrotic stage, leading to severe features and progressive nature of muscular dystrophy.
Collapse
|