1
|
Wang Y, Wang X, Alabdullatif S, Homma ST, Alekseyev YO, Zhou L. Expansion and pathogenic activation of skeletal muscle-resident macrophages in mdx5cv/Ccr2-/- mice. Proc Natl Acad Sci U S A 2025; 122:e2410095122. [PMID: 40067893 PMCID: PMC11929395 DOI: 10.1073/pnas.2410095122] [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: 05/20/2024] [Accepted: 02/10/2025] [Indexed: 03/25/2025] Open
Abstract
Infiltrating macrophages contribute to muscle dystrophic changes in Duchenne muscular dystrophy (DMD). In a DMD mouse model, mdx5cv mice, CC chemokine receptor type 2 (CCR2) deficiency diminishes Ly6Chi macrophage infiltration by blocking blood Ly6Chi inflammatory monocyte recruitment. This is accompanied by transient improvement of muscle damage, fibrosis, and regeneration. The benefit, however, is lost after the expansion of intramuscular Ly6Clo macrophages. To address the mechanisms underlying the Ly6Clo macrophage expansion, we compared mdx5cv/Nur77-/- and mdx5cv/Ccr2-/-/Nur7-/- mice with mdx5cv and mdx5cv/Ccr2-/- mice, respectively, and found no evidence to suggest Ly6Clo monocyte recruitment by dystrophic muscles. Single-cell RNA sequencing analysis and Flt3cre/Rosa26LSL-YFP-based lineage tracing of macrophage origins demonstrated the expansion and pathogenic activation of muscle resident macrophages in CCR2-deficient mdx5cv mice. The expansion was associated with increased cell proliferation, which appeared induced by colony-stimulating factor-1 (CSF-1) derived from fibro/adipogenic progenitors (FAPs). Our study establishes a pathogenic role for skeletal muscle resident macrophages and supports a regulatory role of FAPs in stimulating the expansion of resident macrophages in the DMD mouse model when the inflammatory macrophage infiltration is inhibited.
Collapse
MESH Headings
- Animals
- Receptors, CCR2/genetics
- Receptors, CCR2/metabolism
- Receptors, CCR2/deficiency
- Macrophages/immunology
- Macrophages/metabolism
- Muscle, Skeletal/pathology
- Muscle, Skeletal/metabolism
- Mice
- Muscular Dystrophy, Duchenne/genetics
- Muscular Dystrophy, Duchenne/pathology
- Muscular Dystrophy, Duchenne/immunology
- Mice, Inbred mdx
- Mice, Knockout
- Disease Models, Animal
- Antigens, Ly
Collapse
Affiliation(s)
- Yinhang Wang
- Department of Neurology, Hospital for Special Surgery, New York, NY10021
| | - Xingyu Wang
- Department of Neurology, Hospital for Special Surgery, New York, NY10021
| | - Salam Alabdullatif
- Department of Medicine, Single Cell Sequencing Core Facility, Boston University Chobanian and Avedisian School of Medicine, Boston, MA02118
| | - Sachiko T. Homma
- Department of Neurology, Boston University Chobanian and Avedisian School of Medicine, Boston, MA02118
| | - Yuriy O. Alekseyev
- Department of Pathology and Laboratory Medicine, Boston University Chobanian and Avedisian School of Medicine, Boston, MA02118
| | - Lan Zhou
- Department of Neurology, Hospital for Special Surgery, New York, NY10021
| |
Collapse
|
2
|
K J, Santiago R. Quantitative Structure-Property Relationship Modeling with the Prediction of Physicochemical Properties of Some Novel Duchenne Muscular Dystrophy Drugs. ACS OMEGA 2025; 10:3640-3651. [PMID: 39926532 PMCID: PMC11800030 DOI: 10.1021/acsomega.4c08572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/25/2024] [Accepted: 12/18/2024] [Indexed: 02/11/2025]
Abstract
Duchenne muscular dystrophy is a critical, progressively worsening, and ultimately deadly illness characterized by the deterioration of skeletal muscles, respiratory failure, and heart disease. The pharmaceutical industries are persistently innovating drug design processes to address the rise of infections and effectively treat emerging syndromes or genetically based disorders with the help of quantitative structure-property relationship models. These models are mathematical tools that correlate molecular structures with their physicochemical properties through structural characteristics. Different models can be generated based on the various structural features of the compounds, and topological indices are one such significant structural feature generated from the molecular graph and are key tools used in these models. This study focuses on creating quantitative structure-property relationship models using degree-based topological indices, which are highly effective in quantitative structure-property relationship analysis to explore the diverse physicochemical properties of Duchenne muscular dystrophy drugs with the prediction of properties of a recently approved drug givinostat. Furthermore, the drug discovery and development activities can be accelerated using the developed models to forecast the possible productiveness of novel Duchenne muscular dystrophy treatment drugs.
Collapse
Affiliation(s)
- Jyothish K
- Department of Mathematics,
School of Advanced Sciences, Vellore Institute
of Technology, Vellore 632014, India
| | - Roy Santiago
- Department of Mathematics,
School of Advanced Sciences, Vellore Institute
of Technology, Vellore 632014, India
| |
Collapse
|
3
|
Tomaz da Silva M, Joshi AS, Kumar A. Fibroblast growth factor-inducible 14 regulates satellite cell self-renewal and expansion during skeletal muscle repair. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.06.616900. [PMID: 39803454 PMCID: PMC11722277 DOI: 10.1101/2024.10.06.616900] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Skeletal muscle regeneration in adults is predominantly driven by satellite cells. Loss of satellite cell pool and function leads to skeletal muscle wasting in many conditions and disease states. Here, we demonstrate that the levels of fibroblast growth factor-inducible 14 (Fn14) are increased in satellite cells after muscle injury. Conditional ablation of Fn14 in Pax7-expressing satellite cells drastically reduces their expansion and skeletal muscle regeneration following injury. Fn14 is required for satellite cell self-renewal and proliferation as well as to prevent precocious differentiation. Targeted deletion of Fn14 inhibits Notch signaling but leads to the spurious activation of STAT3 signaling in regenerating skeletal muscle and in cultured muscle progenitor cells. Silencing of STAT3 improves proliferation and inhibits premature differentiation of Fn14-deficient satellite cells. Furthermore, conditional ablation of Fn14 in satellite cells exacerbates myopathy in the mdx mouse model of Duchenne muscular dystrophy (DMD) whereas its overexpression improves the engraftment of exogenous muscle progenitor cells into the dystrophic muscle of mdx mice. Altogether, our study highlights the crucial role of Fn14 in the regulation of satellite cell fate and function and suggests that Fn14 can be a potential molecular target to improve muscle regeneration in muscular disorders.
Collapse
Affiliation(s)
- Meiricris Tomaz da Silva
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Aniket S. Joshi
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| | - Ashok Kumar
- Institute of Muscle Biology and Cachexia, University of Houston College of Pharmacy, Houston, TX, USA
- Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX, USA
| |
Collapse
|
4
|
Cerbin L, Sandhu A, Rosenberg M, Barrett C, Sabzwari R, Garg L, Tumolo A, Tzou W, Varosy P, Von Alvensleben J, Zipse M, Aleong R. Ventricular stimulation in patients with myotonic dystrophy type 1 may not predict future ventricular arrhythmias. Heart Rhythm O2 2024; 5:698-704. [PMID: 39524047 PMCID: PMC11549515 DOI: 10.1016/j.hroo.2024.08.001] [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] [Indexed: 11/16/2024] Open
Abstract
Background Myotonic dystrophy type 1 (DM1) is associated with progressive conduction disease. Furthermore, DM1 patients are at risk ventricular arrhythmias (VAs), although prediction remains difficult. The 2022 Heart Rhythm Expert Consensus Statement gives a IIb recommendation to the use of electrophysiology study (EPS) to risk-stratify patients for VAs. The utility of EPS in predicting the development of VAs, however, has not been explored in this patient population. Objective The study sought to examine the natural history of DM1 patients with positive and negative ventricular stimulation (v-stim) during EPS. Methods Patients with a history of DM1 undergoing EPS with associated v-stim from 2008 to present were retrospectively identified. Results From 2008 to 2022, 26 consecutive DM1 patients presented for EPS with v-stim. Four v-stim protocols were positive for sustained or hemodynamically significant ventricular tachycardia (VT), one of which was induced with 600 doubles, the others with triple extrastimuli. A total of 22 of 26 subjects received a device implant, with 18 receiving permanent pacemakers and 4 implantable cardioverter-defibrillators. All 4 of the patients with positive v-stims underwent ICD implantation. After a mean of 5.7 years of follow-up, 7 patients had sustained VT, 6 of whom had negative v-stims. Of the 4 patients with positive v-stims, only 1 developed sustained VT in follow-up. Other than baseline QT interval at time of EPS, no baseline characteristics were significantly different between patients with and without subsequent VT. Conclusion In this single center, v-stim in DM1 patients did not predict clinical VAs, as a vast majority of DM1 patients who developed VAs had negative v-stims.
Collapse
Affiliation(s)
- Lukasz Cerbin
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Amneet Sandhu
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Division of Cardiology, Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | - Michael Rosenberg
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | | | - Rafay Sabzwari
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Lohit Garg
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Alexis Tumolo
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Wendy Tzou
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Paul Varosy
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
- Division of Cardiology, Rocky Mountain Regional VA Medical Center, Aurora, Colorado
| | | | - Matthew Zipse
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| | - Ryan Aleong
- Division of Cardiology, University of Colorado Anschutz Medical Campus, Aurora, Colorado
| |
Collapse
|
5
|
Okino R, Goda Y, Ono Y. The Hox-based positional memory in muscle stem cells. J Biochem 2024; 176:277-283. [PMID: 39194026 DOI: 10.1093/jb/mvae059] [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: 06/26/2024] [Revised: 08/09/2024] [Accepted: 08/23/2024] [Indexed: 08/29/2024] Open
Abstract
The skeletal muscle is a contractile tissue distributed throughout the body with various anatomical sizes, shapes and functions. In pathological conditions, such as muscular dystrophy, age-related sarcopenia and cancer cachexia, skeletal muscles are not uniformly affected throughout the body. This region-specific vulnerability cannot be fully explained by known physiological classifications, including muscle fiber types. Accumulating evidence indicates that the expression patterns of topographic homeobox (Hox) genes provide a molecular signature of positional memory, reflecting the anatomical locations and embryonic history of muscles and their associated muscle stem cells in adult mice and humans. Hox-based positional memory is not merely a remnant of embryonic development but is expected to be an intrinsic determinant controlling muscle function because recent studies have shown that aberrant Hox genes affect muscle stem cells. In this review, we discuss the concept of Hox-based positional memory, which may offer a new perspective on the region-specific pathophysiology of muscle disorders.
Collapse
Affiliation(s)
- Ryosuke Okino
- Muscle Biology Laboratory, Research Team for Aging Science, Tokyo Metropolitan Institute for Geriatrics and Gerontology (TMIG), 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yuki Goda
- Muscle Biology Laboratory, Research Team for Aging Science, Tokyo Metropolitan Institute for Geriatrics and Gerontology (TMIG), 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yusuke Ono
- Muscle Biology Laboratory, Research Team for Aging Science, Tokyo Metropolitan Institute for Geriatrics and Gerontology (TMIG), 35-2 Sakae-cho, Itabashi-ku, Tokyo, 173-0015, Japan
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics (IMEG), Kumamoto University, 2-2-1 Honjo, Kumamoto, 860-0811, Japan
| |
Collapse
|
6
|
Reynolds RM, Marks SL, Guo LT, Shelton GD, Graham KJ. Feline dystrophin-deficient muscular dystrophy misdiagnosed as Toxoplasma myositis. JFMS Open Rep 2024; 10:20551169241254227. [PMID: 39099732 PMCID: PMC11297512 DOI: 10.1177/20551169241254227] [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] [Indexed: 08/06/2024] Open
Abstract
Case summary A 6-month-old male entire domestic shorthair cat presented for presumptive Toxoplasma myopathy that was non-responsive to antiprotozoal therapy. Clinical features included marked macroglossia, dysphagia, regurgitation, truncal muscle hypertrophy, pelvic limb gait abnormalities and megaoesophagus. Relevant diagnostics included serial creatine kinase activity, cardiac troponin I, fluoroscopic swallow study and routine muscle histopathology. Ultimately, post-mortem histopathology with immunostaining demonstrated markedly decreased or absent staining for the rod and carboxy terminus of dystrophin, confirming a dystrophin-deficient muscular dystrophy (MD). The misdiagnosis of toxoplasmosis was based on an increased IgG titre and muscle histopathology submitted to a local laboratory. Treatment for megaoesophagus included vertical feeding of wet food only, sildenafil and omeprazole. Dysphagia and regurgitation improved moderately. Presumptive hyperaesthesia and muscle pain were managed with anti-inflammatory doses of prednisolone. The patient was ultimately euthanased as a result of progressive MD signs and uraemia at 2 years of age. Relevance and novel information This case report highlights the collective clinical features of MD, as they could be considered pathognomonic for this rare condition and must be differentiated from other myopathies via specific immunostaining of muscle biopsies. This is crucial to obtain a correct and early diagnosis, allowing instigation of potentially valuable treatments. Megaoesophagus is an inconsistent feature in feline MD in addition to the more commonly observed oropharyngeal dysphagia. Management with a canned diet, sildenafil, omeprazole and upright feeding was beneficial with moderate improvement in the frequency of regurgitation. Prednisolone was thought to minimise the presumptive myalgia.
Collapse
Affiliation(s)
| | - Stanley L Marks
- School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Ling T Guo
- Department of Pathology, University of California, San Diego, CA, USA
| | - G. Diane Shelton
- Department of Pathology, University of California, San Diego, CA, USA
| | | |
Collapse
|
7
|
Mavrommatis L, Zaben A, Kindler U, Kienitz MC, Dietz J, Jeong HW, Böhme P, Brand-Saberi B, Vorgerd M, Zaehres H. CRISPR/Cas9 Genome Editing in LGMD2A/R1 Patient-Derived Induced Pluripotent Stem and Skeletal Muscle Progenitor Cells. Stem Cells Int 2023; 2023:9246825. [PMID: 38020204 PMCID: PMC10653971 DOI: 10.1155/2023/9246825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 04/16/2023] [Accepted: 06/08/2023] [Indexed: 12/01/2023] Open
Abstract
Large numbers of Calpain 3 (CAPN3) mutations cause recessive forms of limb-girdle muscular dystrophy (LGMD2A/LGMDR1) with selective atrophy of the proximal limb muscles. We have generated induced pluripotent stem cells (iPSC) from a patient with two mutations in exon 3 and exon 4 at the calpain 3 locus (W130C, 550delA). Two different strategies to rescue these mutations are devised: (i) on the level of LGMD2A-iPSC, we combined CRISPR/Cas9 genome targeting with a FACS and Tet transactivator-based biallelic selection strategy, which resulted in a new functional chimeric exon 3-4 without the two CAPN3 mutations. (ii) On the level of LGMD2A-iPSC-derived CD82+/Pax7+ myogenic progenitor cells, we demonstrate CRISPR/Cas9 mediated rescue of the highly prevalent exon 4 CAPN3 mutation. The first strategy specifically provides isogenic LGMD2A corrected iPSC for disease modelling, and the second strategy can be further elaborated for potential translational approaches.
Collapse
Affiliation(s)
- Lampros Mavrommatis
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular Embryology, 44801 Bochum, Germany
- Ruhr University Bochum, Medical Faculty, Department of Neurology with Heimer Institute for Muscle Research, University Hospital Bergmannsheil, 44789 Bochum, Germany
- Max Planck Institute for Molecular Biomedicine, Department of Cell and Developmental Biology, 48149 Münster, Germany
| | - Abdul Zaben
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular Embryology, 44801 Bochum, Germany
- Ruhr University Bochum, Medical Faculty, Department of Neurology with Heimer Institute for Muscle Research, University Hospital Bergmannsheil, 44789 Bochum, Germany
| | - Urs Kindler
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular Embryology, 44801 Bochum, Germany
| | - Marie-Cécile Kienitz
- Ruhr University Bochum, Medical Faculty, Department of Cellular Physiology, 44801 Bochum, Germany
| | - Julienne Dietz
- Ruhr University Bochum, Medical Faculty, Department of Neurology with Heimer Institute for Muscle Research, University Hospital Bergmannsheil, 44789 Bochum, Germany
- Witten/Herdecke University, Institute of Virology and Microbiology, Department of Human Medicine, Faculty of Health, 58453 Witten, Germany
| | - Hyun-Woo Jeong
- Max Planck Institute for Molecular Biomedicine, Sequencing Core Facility, 48149 Münster, Germany
| | - Pierre Böhme
- Ruhr University Bochum, Department of Psychiatry, Psychotherapy and Preventive Medicine, LWL University Hospital Bochum, 44791 Bochum, Germany
| | - Beate Brand-Saberi
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular Embryology, 44801 Bochum, Germany
| | - Matthias Vorgerd
- Ruhr University Bochum, Medical Faculty, Department of Neurology with Heimer Institute for Muscle Research, University Hospital Bergmannsheil, 44789 Bochum, Germany
| | - Holm Zaehres
- Ruhr University Bochum, Medical Faculty, Institute of Anatomy, Department of Anatomy and Molecular Embryology, 44801 Bochum, Germany
- Max Planck Institute for Molecular Biomedicine, Department of Cell and Developmental Biology, 48149 Münster, Germany
| |
Collapse
|
8
|
Wang X, Zhou L. The multifaceted role of macrophages in homeostatic and injured skeletal muscle. Front Immunol 2023; 14:1274816. [PMID: 37954602 PMCID: PMC10634307 DOI: 10.3389/fimmu.2023.1274816] [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: 08/08/2023] [Accepted: 10/13/2023] [Indexed: 11/14/2023] Open
Abstract
Skeletal muscle is essential for body physical activity, energy metabolism, and temperature maintenance. It has excellent capabilities to maintain homeostasis and to regenerate after injury, which indispensably relies on muscle stem cells, satellite cells (MuSCs). The quiescence, activation, and differentiation of MuSCs are tightly regulated in homeostatic and regenerating muscles. Among the important regulators are intramuscular macrophages, which are functionally heterogeneous with different subtypes present in a spatiotemporal manner to regulate the balance of different MuSC statuses. During chronic injury and aging, intramuscular macrophages often undergo aberrant activation, which in turn disrupts muscle homeostasis and regenerative repair. Growing evidence suggests that the aberrant activation is mainly triggered by altered muscle microenvironment. The trained immunity that affects myeloid progenitors during hematopoiesis may also contribute. Aged immune system may contribute, in part, to the aging-related sarcopenia and compromised skeletal muscle injury repair. As macrophages are actively involved in the progression of many muscle diseases, manipulating their functional activation has become a promising therapeutic approach, which requires comprehensive knowledge of the cellular and molecular mechanisms underlying the diverse activation. To this end, we discuss here the current knowledge of multifaceted role of macrophages in skeletal muscle homeostasis, injury, and repair.
Collapse
Affiliation(s)
- Xingyu Wang
- Department of Neurology, Boston University Chobanian & Avedisian School of Medicine, Boston, MA, United States
| | | |
Collapse
|
9
|
Brown A, Morris B, Kamau JK, Alshudukhi AA, Jama A, Ren H. Automated Image Analysis Pipeline Development to Monitor Disease Progression in Muscular Dystrophy Using Cell Profiler. ARCHIVES OF MICROBIOLOGY & IMMUNOLOGY 2023; 7:178-187. [PMID: 37799294 PMCID: PMC10552673 DOI: 10.26502/ami.936500115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/07/2023]
Abstract
Muscular dystrophies are inherited disorders that are characterized by progressive muscle degeneration. These disorders are caused by mutations in the genes encoding structural elements within the muscle, which leads to increased vulnerability to mechanical stress and sarcolemma damage. Although myofibers have the capacity to regenerate, the newly formed myofibers still harbor genetic mutation, which induces continuous cycles of muscle fiber death and regeneration. This repeated cycling is accompanied by an inflammatory response which eventually provokes excessive fibrotic deposition. The histopathological changes in skeletal muscle tissue are central to the disease pathogenesis. Analysis of muscle histopathology is the gold standard for monitoring muscle health and disease progression. However, manual, or semi-manual quantification methods, are not only immensely tedious but can be subjective. Here, we present four image analysis pipelines built in CellProfiler which enable users without a background in computer vision or programming to quantitatively analyze biological images. These image analysis pipelines are designed to quantify skeletal muscle histopathological staining for membrane damage, the abundance and size distribution of regenerating muscle fibers, inflammation via quantification of CD68+ M1 macrophages, and collagen deposition. Additionally, we discuss methods to address common errors associated with the quantification of microscopy images. These automated tools can not only improve workflow efficiency but can provide a better understanding of the histopathological progression of muscular dystrophy.
Collapse
Affiliation(s)
- Alexandra Brown
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| | - Brooklyn Morris
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| | - John Karanja Kamau
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| | - Abdullah A Alshudukhi
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| | - Abdulrahman Jama
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| | - Hongmei Ren
- Department of Biochemistry and Molecular Biology, Wright State University, Dayton, OH, USA
| |
Collapse
|
10
|
Roy A, Koike TE, Joshi AS, Tomaz da Silva M, Mathukumalli K, Wu M, Kumar A. Targeted regulation of TAK1 counteracts dystrophinopathy in a DMD mouse model. JCI Insight 2023; 8:e164768. [PMID: 37071470 PMCID: PMC10322678 DOI: 10.1172/jci.insight.164768] [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: 08/24/2022] [Accepted: 04/12/2023] [Indexed: 04/19/2023] Open
Abstract
Muscular dystrophies make up a group of genetic neuromuscular disorders that involve severe muscle wasting. TGF-β-activated kinase 1 (TAK1) is an important signaling protein that regulates cell survival, growth, and inflammation. TAK1 has been recently found to promote myofiber growth in the skeletal muscle of adult mice. However, the role of TAK1 in muscle diseases remains poorly understood. In the present study, we have investigated how TAK1 affects the progression of dystrophic phenotype in the mdx mouse model of Duchenne muscular dystrophy (DMD). TAK1 is highly activated in the dystrophic muscle of mdx mice during the peak necrotic phase. While targeted inducible inactivation of TAK1 inhibits myofiber injury in young mdx mice, it results in reduced muscle mass and contractile function. TAK1 inactivation also causes loss of muscle mass in adult mdx mice. By contrast, forced activation of TAK1 through overexpression of TAK1 and TAB1 induces myofiber growth without having any deleterious effect on muscle histopathology. Collectively, our results suggest that TAK1 is a positive regulator of skeletal muscle mass and that targeted regulation of TAK1 can suppress myonecrosis and ameliorate disease progression in DMD.
Collapse
|
11
|
Leiva D, Sepúlveda C, Toledo LD. Alteraciones del habla y deglución en pacientes con distrofia muscular: una revisión sistemática. REVISTA DE INVESTIGACIÓN EN LOGOPEDIA 2023. [DOI: 10.5209/rlog.83585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
La distrofia muscular corresponde a un grupo heterogéneo de alteraciones musculares de origen genético. El propósito de esta revisión fue describir las principales alteraciones del habla y deglución que presentan los pacientes con distrofia muscular. Se realizó una búsqueda electrónica de artículos relevantes en el área, los cuales incluyeron en su descripción pacientes con distrofia muscular asociadas a trastornos del habla y/o deglución. Las bases de datos revisadas fueron EMBASE, CINAHL, PubMed, PsycInfo, Web of Science y Scopus. Se encontraron 15 estudios que cumplieron con los criterios de inclusión, involucrando un total de 526 participantes con un promedio de edad de 43,09 años. 12/15 estudios incluyeron medidas de deglución y/o alimentación y 3/15 incluyeron evaluación del habla. La revisión evidencia gran variabilidad en los instrumentos utilizados para describir las alteraciones del habla y deglución. En las distrofias musculares incluidas en la presente revisión se observó alteración principalmente en la etapa faríngea y dificultad en la formación del bolo asociado a alteraciones en la oclusión y fuerza muscular. El habla es un parámetro poco estudiado en este tipo de condición.
Collapse
|
12
|
Barboni MTS, Joachimsthaler A, Roux MJ, Nagy ZZ, Ventura DF, Rendon A, Kremers J, Vaillend C. Retinal dystrophins and the retinopathy of Duchenne muscular dystrophy. Prog Retin Eye Res 2022:101137. [DOI: 10.1016/j.preteyeres.2022.101137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 10/25/2022] [Accepted: 11/03/2022] [Indexed: 11/21/2022]
|
13
|
Oliveira-Santos A, Dagda M, Burkin DJ. Sunitinib inhibits STAT3 phosphorylation in cardiac muscle and prevents cardiomyopathy in the mdx mouse model of Duchenne muscular dystrophy. Hum Mol Genet 2022; 31:2358-2369. [PMID: 35157045 PMCID: PMC9307308 DOI: 10.1093/hmg/ddac042] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/31/2022] [Accepted: 02/09/2022] [Indexed: 11/14/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-linked genetic disorder affecting approximately 1 in 5000 male births worldwide. DMD is caused by mutations in the dystrophin gene. Dystrophin is essential for maintaining muscle cell membrane integrity and stability by linking the cytoskeleton to the extracellular matrix, which protects myofibers from contraction-induced damage. Loss of dystrophin leads to mechanically induced skeletal and cardiac muscle damage. Although the disease is not evident in DMD patients at birth, muscular dystrophy rapidly progresses and results in respiratory and cardiac muscle failure as early as the teenage years. Premature death in DMD patients is due to cardiac arrhythmias and left ventricular dysfunction. Currently, there is no effective treatment for DMD-related cardiac failure. Recently, we have shown that a Food and Drug Administration-approved small molecule, sunitinib, a multi-targeted tyrosine kinase inhibitor can mitigate skeletal muscle disease through an increase in myogenic capacity, cell membrane integrity, and improvement of skeletal muscle function via regulation of STAT3-related signaling pathway. Chronic activation of STAT3 has been shown to promote cardiac hypertrophy and failure. In this study, we examined the effects of long-term sunitinib treatment on cardiac pathology and function. Our results showed sunitinib treatment reduced STAT3 phosphorylation in the heart muscle of mdx mice, improved cardiac electrical function, increased cardiac output and stroke volume, decreased ventricular hypertrophy, reduced cardiomyocytes membrane damage, fibrotic tissue deposition and slightly decreased cardiac inflammation. Together, our studies support the idea that sunitinib could serve as a novel treatment to slow cardiomyopathy progression in DMD. One Sentence Summary In this study, we determined if sunitinib, a Food and Drug Administration-approved drug, could reduce the pathology and improve cardiac function in an animal model for DMD.
Collapse
Affiliation(s)
- Ariany Oliveira-Santos
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno NV 89557, USA
| | - Marisela Dagda
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno NV 89557, USA
| | - Dean J Burkin
- Department of Pharmacology, University of Nevada Reno, School of Medicine, Center for Molecular Medicine, Reno NV 89557, USA
| |
Collapse
|
14
|
Chen L, Xu B, Liu W, Wang D, Xu J, Mao X, Xiao M, Zhou J, Xiao Y. CAR T-Cell Therapy for Relapsed/Refractory Diffuse Large B-Cell Lymphoma with Progressive Muscular Dystrophy: A Case Report. Onco Targets Ther 2022; 15:361-366. [PMID: 35422632 PMCID: PMC9005144 DOI: 10.2147/ott.s352760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/01/2022] [Indexed: 11/23/2022] Open
Abstract
Muscular dystrophies are a heterogeneous group of genetically inherited degenerative disorders defined by dystrophic features on pathological assessment of muscle biopsy specimens. Muscular dystrophies and lymphoma are not common concomitant diseases. Chimeric antigen receptor (CAR) T-cell immunotherapy for lymphoma patients with inherited degenerative diseases, such as muscular dystrophies, has not been previously reported. We report a relapsed/refractory diffuse large B-cell lymphoma (DLBCL) patient with progressive muscular dystrophy (PMD) characterized by progressive muscle weakness that affected the limb, axial and facial muscles. He was identified to be a germline DYSF p.R204* homozygous mutation carrier. The patient received a murine monoclonal anti-CD19 and anti-CD22 CAR T-cell “cocktail” and suffered from a mild case of grade 1 cytokine release syndrome (CRS). One month after the CAR T-cell infusion, he achieved complete remission of his lymphoma without minimal residual disease (MRD), as assessed by radiography. One year after the infusion, the Deauville score was stable at 1. Currently, patient has been in remission for over three years after receiving anti-CD19 and anti-CD22 CAR T-cell therapy. This case provides evidence for the use of CAR T-cell therapy in lymphoma patients with inherited degenerative disorders. Achieving remission of the lymphoma and subsequent administration of γ-globulin as well as zoledronic acid reduced the muscular dystrophy symptoms.
Collapse
Affiliation(s)
- Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Bin Xu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Wanying Liu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Di Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Jinhuan Xu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Xia Mao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Min Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Jianfeng Zhou
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
| | - Yi Xiao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People’s Republic of China
- Correspondence: Yi Xiao; Jianfeng Zhou, Email ;
| |
Collapse
|
15
|
Abstract
PURPOSE OF REVIEW This review highlights the key studies investigating various types of biomarkers in Duchenne muscular dystrophy (DMD). RECENT FINDINGS Several proteomic and metabolomic studies have been undertaken in both human DMD patients and animal models of DMD that have identified potential biomarkers in DMD. Although there have been a number of proteomic and metabolomic studies that have identified various potential biomarkers in DMD, more definitive studies still need to be undertaken in DMD patients to firmly correlate these biomarkers with diagnosis, disease progression, and monitoring the effects of novel treatment strategies being developed.
Collapse
Affiliation(s)
- Theo Lee-Gannon
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
| | - Xuan Jiang
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Tara C Tassin
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Pradeep P A Mammen
- Division of Cardiology, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- Department of Internal Medicine, UT Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas, TX, 75390, USA.
- UT Southwestern Senator Paul D. Wellstone Muscular Dystrophy Specialized Research Center, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Heart Failure, Ventricular Assist Device & Heart Transplant Program, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
- Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, TX, 75390, USA.
| |
Collapse
|
16
|
Saclier M, Angelini G, Bonfanti C, Mura G, Temponi G, Messina G. Selective ablation of Nfix in macrophages attenuates muscular dystrophy by inhibiting fibro-adipogenic progenitor-dependent fibrosis. J Pathol 2022; 257:352-366. [PMID: 35297529 PMCID: PMC9322546 DOI: 10.1002/path.5895] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 02/24/2022] [Accepted: 03/15/2022] [Indexed: 11/10/2022]
Abstract
Muscular dystrophies are genetic diseases characterized by chronic inflammation and fibrosis. Macrophages are immune cells that sustain muscle regeneration upon acute injury but seem deleterious in the context of chronic muscle injury such as in muscular dystrophies. Here, we observed that the number of macrophages expressing the transcription factor Nfix increases in two distinct mouse models of muscular dystrophies. We showed that the deletion of Nfix in macrophages in dystrophic mice delays the establishment of fibrosis and muscle wasting, and increases grasp force. Macrophages lacking Nfix expressed more TNFα and less TGFβ1, thus promoting apoptosis of fibro‐adipogenic progenitors. Moreover, pharmacological treatment of dystrophic mice with a ROCK inhibitor accelerated fibrosis through the increase of Nfix expression by macrophages. Thus, we have identified Nfix as a macrophage profibrotic factor in muscular dystrophies, whose inhibition could be a therapeutic route to reduce severity of the dystrophic disease. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | | | - Chiara Bonfanti
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giada Mura
- Department of Biosciences, University of Milan, Milan, Italy
| | - Giulia Temponi
- Department of Biosciences, University of Milan, Milan, Italy
| | | |
Collapse
|
17
|
Ansaripour A, Roehrich K, Mashayekhi A, Wanjala M, Noel S, Hemami MR, Murray A, Javanbakht M. Budget Impact of the Vest™ High Frequency Chest Wall Oscillation System for Managing Airway Clearance in Patients with Complex Neurological Disorders: A US Healthcare Payers' Perspective Analysis. PHARMACOECONOMICS - OPEN 2022; 6:169-178. [PMID: 34468966 PMCID: PMC8864050 DOI: 10.1007/s41669-021-00299-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 08/08/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE As a recognized therapy to improve airway clearance, high-frequency chest wall oscillation (HFCWO) is used to manage reduced vital capacity in patients with complex neuromuscular disorders (cNMD). This study aimed to investigate the budget impact of HFCWO versus chest wall physical therapy (CWPT) from a US-commercial payer perspective. METHODS In combination with a previously developed cost-effectiveness model, a budget impact model was developed to evaluate the incremental budgetary impact associated with introducing a HFCWO device over a 5-year time horizon. The model compared the cost implications associated with the commonly used CWPT procedure, as the current scenario, with a new scenario consisting of 80% of market share for HFCWO. The resource use and costs included in the analyses were costs associated with the HFCWO device (Vest™ System) and its consumables, patient training, and medical services such as hospitalization, medications, emergency room, and outpatient visits. The primary outcome measures included total and incremental budgetary impact per member per year (PMPY). RESULTS In a hypothetical plan of 1,000,000 members (men: 49.2%), 2099 patients with cNMD were estimated to be eligible to receive airway clearance services over 5 years. The new scenario (HFCWO and CWPT [US$24 PMPY]) was cost-saving compared with the current scenario (CWPT only [$34 PMPY]) with a cost reduction of US$9.46 PMPY. The model estimated a net cost-saving of US$1,594,131 and US$9,591,343 over 1 and 5 years, respectively. CONCLUSION This study suggests the HFCWO technique to manage the reduction in vital capacity in patients with cNMD would lead to favorable budget impact results.
Collapse
Affiliation(s)
- Amir Ansaripour
- Optimax Access, Gustav Mahlerplein 2, 1082 MA, Amsterdam, The Netherlands.
| | | | - Atefeh Mashayekhi
- Optimax Access, University of Southampton Science Park, Chilworth, Hampshire, UK
| | | | | | | | | | - Mehdi Javanbakht
- Optimax Access, University of Southampton Science Park, Chilworth, Hampshire, UK
- Device Access, University of Southampton Science Park, Chilworth, Hampshire, UK
| |
Collapse
|
18
|
Goto A, Kokabu S, Dusadeemeelap C, Kawaue H, Matsubara T, Tominaga K, Addison WN. Tongue Muscle for the Analysis of Head Muscle Regeneration Dynamics. J Dent Res 2022; 101:962-971. [PMID: 35193429 DOI: 10.1177/00220345221075966] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Tongue muscle damage impairs speaking and eating, thereby degrading overall health and quality of life. Skeletal muscles of the body are diverse in embryonic origin, anatomic location, and gene expression profiles. Responses to disease, atrophy, aging, or drugs vary among different muscles. Currently, most muscle studies are focused on limb muscles and the tongue is neglected. The regenerative ability of tongue muscle remains unknown, and thus there is need for tongue muscle research models. Here, we present a comprehensive characterization of the spatiotemporal dynamics in a mouse model of tongue muscle regeneration and establish a method for the isolation of primary tongue-derived satellite cells. We compare and contrast our observations with the tibialis anterior (TA) limb muscle. Acute injury was induced by intramuscular injection of cardiotoxin, a cytolytic agent, and examined at multiple timepoints. Initially, necrotic myofibers with fragmented sarcoplasm became infiltrated with inflammatory cells. Concomitantly, satellite cells expanded rapidly. Seven days postinjury, regenerated myofibers with centralized nuclei appeared. Full regeneration, as well as an absence of fibrosis, was evident 21 d postinjury. Primary tongue-derived satellite cells were isolated by enzymatic separation of tongue epithelium from mesenchyme followed by magnetic-activated cell sorting. We observed that tongue displays an efficient regenerative response similar to TA but with slightly faster kinetics. In vitro, tongue-derived satellite cells differentiated robustly into mature myotubes with spontaneous contractile behavior and myogenic marker expression. Comparison of gene expression signatures between tongue and TA-derived satellite cells revealed differences in the expression of positional-identity genes, including the HOX family. In conclusion, we have established a model for tongue regeneration useful for investigations of orofacial muscle biology. Furthermore, we showed that tongue is a viable source of satellite cells with unique properties and inherited positional memory.
Collapse
Affiliation(s)
- A Goto
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu, Fukuoka, Japan.,Division of Oral and Maxillofacial Surgery, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - S Kokabu
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - C Dusadeemeelap
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - H Kawaue
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - T Matsubara
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - K Tominaga
- Division of Oral and Maxillofacial Surgery, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| | - W N Addison
- Division of Molecular Signaling and Biochemistry, Kyushu Dental University, Kitakyushu, Fukuoka, Japan
| |
Collapse
|
19
|
Roy A, Tomaz da Silva M, Bhat R, Bohnert KR, Iwawaki T, Kumar A. The IRE1/XBP1 signaling axis promotes skeletal muscle regeneration through a cell non-autonomous mechanism. eLife 2021; 10:e73215. [PMID: 34812145 PMCID: PMC8635982 DOI: 10.7554/elife.73215] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Accepted: 11/22/2021] [Indexed: 12/12/2022] Open
Abstract
Skeletal muscle regeneration is regulated by coordinated activation of multiple signaling pathways. The unfolded protein response (UPR) is a major mechanism that detects and alleviates protein-folding stresses in the endoplasmic reticulum. However, the role of individual arms of the UPR in skeletal muscle regeneration remain less understood. In the present study, we demonstrate that IRE1α (also known as ERN1) and its downstream target, XBP1, are activated in skeletal muscle of mice upon injury. Myofiber-specific ablation of IRE1α or XBP1 in mice diminishes skeletal muscle regeneration that is accompanied with reduced number of satellite cells. Ex vivo cultures of myofiber explants demonstrate that ablation of IRE1α reduces the proliferative capacity of myofiber-associated satellite cells. Myofiber-specific ablation of IRE1α dampens Notch signaling and canonical NF-κB pathway in skeletal muscle of adult mice. Finally, targeted ablation of IRE1α also reduces Notch signaling, abundance of satellite cells, and skeletal muscle regeneration in the mdx mice, a model of Duchenne muscular dystrophy. Collectively, our experiments suggest that the IRE1α-mediated signaling promotes muscle regeneration through augmenting the proliferation of satellite cells in a cell non-autonomous manner.
Collapse
Affiliation(s)
- Anirban Roy
- Department of Pharmacological and Pharmaceutical Sciences, University of HoustonHoustonUnited States
| | - Meiricris Tomaz da Silva
- Department of Pharmacological and Pharmaceutical Sciences, University of HoustonHoustonUnited States
| | - Raksha Bhat
- Department of Pharmacological and Pharmaceutical Sciences, University of HoustonHoustonUnited States
| | - Kyle R Bohnert
- Kinesiology Department, St Ambrose UniversityDavenportUnited States
| | - Takao Iwawaki
- Division of Cell Medicine, Department of Life Science, Medical Research Institute, Kanazawa Medical UniversityUchinadaJapan
| | - Ashok Kumar
- Department of Pharmacological and Pharmaceutical Sciences, University of HoustonHoustonUnited States
| |
Collapse
|
20
|
Kim H, Kwon Y, Zhu C, Wu F, Kwon S, Yeo W, Choo HJ. Real-Time Functional Assay of Volumetric Muscle Loss Injured Mouse Masseter Muscles via Nanomembrane Electronics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:e2101037. [PMID: 34218527 PMCID: PMC8425913 DOI: 10.1002/advs.202101037] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/28/2021] [Indexed: 05/11/2023]
Abstract
Skeletal muscle has a remarkable regeneration capacity to recover its structure and function after injury, except for the traumatic loss of critical muscle volume, called volumetric muscle loss (VML). Although many extremity VML models have been conducted, craniofacial VML has not been well-studied due to unavailable in vivo assay tools. Here, this paper reports a wireless, noninvasive nanomembrane system that integrates skin-wearable printed sensors and electronics for real-time, continuous monitoring of VML on craniofacial muscles. The craniofacial VML model, using biopsy punch-induced masseter muscle injury, shows impaired muscle regeneration. To measure the electrophysiology of small and round masseter muscles of active mice during mastication, a wearable nanomembrane system with stretchable graphene sensors that can be laminated to the skin over target muscles is utilized. The noninvasive system provides highly sensitive electromyogram detection on masseter muscles with or without VML injury. Furthermore, it is demonstrated that the wireless sensor can monitor the recovery after transplantation surgery for craniofacial VML. Overall, the presented study shows the enormous potential of the masseter muscle VML injury model and wearable assay tool for the mechanism study and the therapeutic development of craniofacial VML.
Collapse
Affiliation(s)
- Hojoong Kim
- George W. Woodruff School of Mechanical EngineeringCollege of EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- Center for Human‐Centric Interfaces and EngineeringInstitute for Electronics and NanotechnologyGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Young‐Tae Kwon
- Department for Metal PowderKorea Institute of Materials ScienceChangwon51508South Korea
| | - Carol Zhu
- Department of Cell BiologySchool of MedicineEmory UniversityAtlantaGA30322USA
| | - Fang Wu
- Department of Cell BiologySchool of MedicineEmory UniversityAtlantaGA30322USA
| | - Shinjae Kwon
- George W. Woodruff School of Mechanical EngineeringCollege of EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- Center for Human‐Centric Interfaces and EngineeringInstitute for Electronics and NanotechnologyGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Woon‐Hong Yeo
- George W. Woodruff School of Mechanical EngineeringCollege of EngineeringGeorgia Institute of TechnologyAtlantaGA30332USA
- Center for Human‐Centric Interfaces and EngineeringInstitute for Electronics and NanotechnologyGeorgia Institute of TechnologyAtlantaGA30332USA
- Wallace H. Coulter Department of Biomedical EngineeringParker H. Petit Institute for Bioengineering and BiosciencesInstitute for MaterialsNeural Engineering CenterInstitute for Robotics and Intelligent MachinesGeorgia Institute of TechnologyAtlantaGA30332USA
| | - Hyojung J. Choo
- Department of Cell BiologySchool of MedicineEmory UniversityAtlantaGA30322USA
| |
Collapse
|
21
|
Barefield DY, Sell JJ, Tahtah I, Kearns SD, McNally EM, Demonbreun AR. Loss of dysferlin or myoferlin results in differential defects in excitation-contraction coupling in mouse skeletal muscle. Sci Rep 2021; 11:15865. [PMID: 34354129 PMCID: PMC8342512 DOI: 10.1038/s41598-021-95378-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 07/26/2021] [Indexed: 11/25/2022] Open
Abstract
Muscular dystrophies are disorders characterized by progressive muscle loss and weakness that are both genotypically and phenotypically heterogenous. Progression of muscle disease arises from impaired regeneration, plasma membrane instability, defective membrane repair, and calcium mishandling. The ferlin protein family, including dysferlin and myoferlin, are calcium-binding, membrane-associated proteins that regulate membrane fusion, trafficking, and tubule formation. Mice lacking dysferlin (Dysf), myoferlin (Myof), and both dysferlin and myoferlin (Fer) on an isogenic inbred 129 background were previously demonstrated that loss of both dysferlin and myoferlin resulted in more severe muscle disease than loss of either gene alone. Furthermore, Fer mice had disordered triad organization with visibly malformed transverse tubules and sarcoplasmic reticulum, suggesting distinct roles of dysferlin and myoferlin. To assess the physiological role of disorganized triads, we now assessed excitation contraction (EC) coupling in these models. We identified differential abnormalities in EC coupling and ryanodine receptor disruption in flexor digitorum brevis myofibers isolated from ferlin mutant mice. We found that loss of dysferlin alone preserved sensitivity for EC coupling and was associated with larger ryanodine receptor clusters compared to wildtype myofibers. Loss of myoferlin alone or together with a loss of dysferlin reduced sensitivity for EC coupling, and produced disorganized and smaller ryanodine receptor cluster size compared to wildtype myofibers. These data reveal impaired EC coupling in Myof and Fer myofibers and slightly potentiated EC coupling in Dysf myofibers. Despite high homology, dysferlin and myoferlin have differential roles in regulating sarcotubular formation and maintenance resulting in unique impairments in calcium handling properties.
Collapse
Affiliation(s)
- David Y Barefield
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA. .,Department of Cell and Molecular Physiology, Loyola University Chicago, 2160 S. 1st Ave, Maywood, IL, 60153, USA.
| | - Jordan J Sell
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Ibrahim Tahtah
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Samuel D Kearns
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Feinberg School of Medicine, Northwestern University, 303 E Superior Lurie 5-500, Chicago, IL, 60611, USA. .,Department of Pharmacology, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA. .,Center for Genetic Medicine, Northwestern University, 303 E Superior Lurie 5-512, Chicago, IL, 60611, USA.
| |
Collapse
|
22
|
Quantitative Muscle MRI in Patients with Neuromuscular Diseases-Association of Muscle Proton Density Fat Fraction with Semi-Quantitative Grading of Fatty Infiltration and Muscle Strength at the Thigh Region. Diagnostics (Basel) 2021; 11:diagnostics11061056. [PMID: 34201303 PMCID: PMC8230029 DOI: 10.3390/diagnostics11061056] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Revised: 06/05/2021] [Accepted: 06/06/2021] [Indexed: 11/17/2022] Open
Abstract
(1) Background and Purpose: The skeletal muscles of patients suffering from neuromuscular diseases (NMD) are affected by atrophy, hypertrophy, fatty infiltration, and edematous changes. Magnetic resonance imaging (MRI) is an important tool for diagnosis and monitoring. Concerning fatty infiltration, T1-weighted or T2-weighted DIXON turbo spin echo (TSE) sequences enable a qualitative assessment of muscle involvement. To achieve higher comparability, semi-quantitative grading scales, such as the four-point Mercuri scale, are commonly applied. However, the evaluation remains investigator-dependent. Therefore, effort is being invested to develop quantitative MRI techniques for determination of imaging markers such as the proton density fat fraction (PDFF). The present work aims to assess the diagnostic value of PDFF in correlation to Mercuri grading and clinically determined muscle strength in patients with myotonic dystrophy type 2 (DM2), limb girdle muscular dystrophy type 2A (LGMD2A), and adult Pompe disease. (2) Methods: T2-weighted two-dimensional (2D) DIXON TSE and chemical shift encoding-based water-fat MRI were acquired in 13 patients (DM2: n = 5; LGMD2A: n = 5; Pompe disease: n = 3). Nine different thigh muscles were rated in all patients according to the Mercuri grading and segmented to extract PDFF values. Muscle strength was assessed according to the British Medical Research Council (BMRC) scale. For correlation analyses between Mercuri grading, muscle strength, and PDFF, the Spearman correlation coefficient (rs) was computed. (3) Results: Mean PDFF values ranged from 7% to 37% in adults with Pompe disease and DM2 and up to 79% in LGMD2A patients. In all three groups, a strong correlation of the Mercuri grading and PDFF values was observed for almost all muscles (rs > 0.70, p < 0.05). PDFF values correlated significantly to muscle strength for muscle groups responsible for knee flexion (rs = -0.80, p < 0.01). (4) Conclusion: In the small, investigated patient cohort, PDFF offers similar diagnostic precision as the clinically established Mercuri grading. Based on these preliminary data, PDFF could be further considered as an MRI-based biomarker in the assessment of fatty infiltration of muscle tissue in NMD. Further studies with larger patient cohorts are needed to advance PDFF as an MRI-based biomarker in NMD, with advantages such as its greater dynamic range, enabling the assessment of subtler changes, the amplified objectivity, and the potential of direct correlation to muscle function for selected muscles.
Collapse
|
23
|
Yoshioka K, Nagahisa H, Miura F, Araki H, Kamei Y, Kitajima Y, Seko D, Nogami J, Tsuchiya Y, Okazaki N, Yonekura A, Ohba S, Sumita Y, Chiba K, Ito K, Asahina I, Ogawa Y, Ito T, Ohkawa Y, Ono Y. Hoxa10 mediates positional memory to govern stem cell function in adult skeletal muscle. SCIENCE ADVANCES 2021; 7:7/24/eabd7924. [PMID: 34108202 PMCID: PMC8189581 DOI: 10.1126/sciadv.abd7924] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 04/21/2021] [Indexed: 05/04/2023]
Abstract
Muscle stem cells (satellite cells) are distributed throughout the body and have heterogeneous properties among muscles. However, functional topographical genes in satellite cells of adult muscle remain unidentified. Here, we show that expression of Homeobox-A (Hox-A) cluster genes accompanied with DNA hypermethylation of the Hox-A locus was robustly maintained in both somite-derived muscles and their associated satellite cells in adult mice, which recapitulates their embryonic origin. Somite-derived satellite cells were clearly separated from cells derived from cranial mesoderm in Hoxa10 expression. Hoxa10 inactivation led to genomic instability and mitotic catastrophe in somite-derived satellite cells in mice and human. Satellite cell-specific Hoxa10 ablation in mice resulted in a decline in the regenerative ability of somite-derived muscles, which were unobserved in cranial mesoderm-derived muscles. Thus, our results show that Hox gene expression profiles instill the embryonic history in satellite cells as positional memory, potentially modulating region-specific pathophysiology in adult muscles.
Collapse
Affiliation(s)
- Kiyoshi Yoshioka
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Hiroshi Nagahisa
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
| | - Fumihito Miura
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Hiromitsu Araki
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yasutomi Kamei
- Graduate School of Life and Environmental Sciences, Kyoto Prefectural University, Kyoto 606-8522, Japan
| | - Yasuo Kitajima
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Daiki Seko
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Jumpei Nogami
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yoshifumi Tsuchiya
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Narihiro Okazaki
- Department of Orthopaedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Akihiko Yonekura
- Department of Orthopaedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Seigo Ohba
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yoshinori Sumita
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Ko Chiba
- Department of Orthopaedic Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Kosei Ito
- Department of Molecular Bone Biology, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8501, Japan
| | - Yoshihiro Ogawa
- Department of Medicine and Bioregulatory Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takashi Ito
- Department of Biochemistry, Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Yasuyuki Ohkawa
- Division of Transcriptomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto 860-0811, Japan.
- Musculoskeletal Molecular Biology Research Group, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki 852-8588, Japan
- Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Kumamoto 860-0811, Japan
| |
Collapse
|
24
|
Farini A, Sitzia C, Villa C, Cassani B, Tripodi L, Legato M, Belicchi M, Bella P, Lonati C, Gatti S, Cerletti M, Torrente Y. Defective dystrophic thymus determines degenerative changes in skeletal muscle. Nat Commun 2021; 12:2099. [PMID: 33833239 PMCID: PMC8032677 DOI: 10.1038/s41467-021-22305-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 02/24/2021] [Indexed: 02/02/2023] Open
Abstract
In Duchenne muscular dystrophy (DMD), sarcolemma fragility and myofiber necrosis produce cellular debris that attract inflammatory cells. Macrophages and T-lymphocytes infiltrate muscles in response to damage-associated molecular pattern signalling and the release of TNF-α, TGF-β and interleukins prevent skeletal muscle improvement from the inflammation. This immunological scenario was extended by the discovery of a specific response to muscle antigens and a role for regulatory T cells (Tregs) in muscle regeneration. Normally, autoimmunity is avoided by autoreactive T-lymphocyte deletion within thymus, while in the periphery Tregs monitor effector T-cells escaping from central regulatory control. Here, we report impairment of thymus architecture of mdx mice together with decreased expression of ghrelin, autophagy dysfunction and AIRE down-regulation. Transplantation of dystrophic thymus in recipient nude mice determine the up-regulation of inflammatory/fibrotic markers, marked metabolic breakdown that leads to muscle atrophy and loss of force. These results indicate that involution of dystrophic thymus exacerbates muscular dystrophy by altering central immune tolerance.
Collapse
Affiliation(s)
- Andrea Farini
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Clementina Sitzia
- Residency Program in Clinical Pathology and Clinical Biochemistry, Università degli Studi di Milano, Milan, Italy
| | - Chiara Villa
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Barbara Cassani
- Consiglio Nazionale delle Ricerche-Istituto di Ricerca Genetica e Biomedica (CNR-IRGB), Milan Unit, Milan, Italy
- IRCCS Humanitas clinical and research center, Rozzano, 20089, Milan, Italy
| | - Luana Tripodi
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Mariella Legato
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Marzia Belicchi
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Pamela Bella
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Caterina Lonati
- Center for Surgical Research, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Gatti
- Center for Surgical Research, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Massimiliano Cerletti
- UCL Research Department for Surgical Biotechnology, University College London, London, UK
- UCL Institute for Immunity and Transplantation, University College London, London, UK
| | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy.
| |
Collapse
|
25
|
Goyal M, Gupta A, Agarwal K, Kapoor S, Kumar S. Duchenne muscular dystrophy: Genetic and clinical profile in the population of Rajasthan, India. Ann Indian Acad Neurol 2021; 24:873-878. [PMID: 35359537 PMCID: PMC8965960 DOI: 10.4103/aian.aian_126_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/16/2021] [Accepted: 03/18/2021] [Indexed: 11/04/2022] Open
Abstract
Background: Materials and Methods: Results: Conclusions:
Collapse
|
26
|
Yoshioka K, Kitajima Y, Seko D, Tsuchiya Y, Ono Y. The body region specificity in murine models of muscle regeneration and atrophy. Acta Physiol (Oxf) 2021; 231:e13553. [PMID: 32875719 PMCID: PMC7757168 DOI: 10.1111/apha.13553] [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: 07/03/2020] [Revised: 08/15/2020] [Accepted: 08/26/2020] [Indexed: 01/06/2023]
Abstract
AIM Skeletal muscles are distributed throughout the body, presenting a variety of sizes, shapes and functions. Here, we examined whether muscle regeneration and atrophy occurred homogeneously throughout the body in mouse models. METHODS Acute muscle regeneration was induced by a single intramuscular injection of cardiotoxin in adult mice. Chronic muscle regeneration was assessed in mdx mice. Muscle atrophy in different muscles was evaluated by cancer cachexia, ageing and castration mouse models. RESULTS We found that, in the cardiotoxin-injected acute muscle injury model, head muscles slowly regenerated, while limb muscles exhibited a rapid regeneration and even overgrowth. This overgrowth was also observed in limb muscles alone (but not in head muscles) in mdx mice as chronic injury models. We described the body region-specific decline in the muscle mass in muscle atrophy models: cancer cachexia-induced, aged and castrated mice. The positional identities, including gene expression profiles and hormone sensitivity, were robustly preserved in the ectopically engrafted satellite cell-derived muscles in the castrated model. CONCLUSION Our results indicate that positional identities in muscles should be considered for the development of efficient regenerative therapies for muscle weakness, such as muscular dystrophy and age-related sarcopenia.
Collapse
Affiliation(s)
- Kiyoshi Yoshioka
- Department of Muscle Development and Regeneration Institute of Molecular Embryology and Genetics Kumamoto University Kumamoto Japan
| | - Yasuo Kitajima
- Department of Muscle Development and Regeneration Institute of Molecular Embryology and Genetics Kumamoto University Kumamoto Japan
| | - Daiki Seko
- Department of Muscle Development and Regeneration Institute of Molecular Embryology and Genetics Kumamoto University Kumamoto Japan
| | - Yoshifumi Tsuchiya
- Department of Muscle Development and Regeneration Institute of Molecular Embryology and Genetics Kumamoto University Kumamoto Japan
| | - Yusuke Ono
- Department of Muscle Development and Regeneration Institute of Molecular Embryology and Genetics Kumamoto University Kumamoto Japan
| |
Collapse
|
27
|
Nassoro DD, Torres L, Marando R, Mboma L, Mushi S, Habakkuk Mwakyula I. A child with duchenne muscular dystrophy: A case report of a rare diagnosis among Africans. Clin Case Rep 2020; 8:2654-2660. [PMID: 33363799 PMCID: PMC7752564 DOI: 10.1002/ccr3.3254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 01/14/2023] Open
Abstract
In Africa, lack of awareness and low index of suspicion of rare diseases like dystrophinopathies, directly or indirectly, contributes to the increased morbidity and mortality. Therefore, even though the data on prevalence is limited, we need to have a high degree of suspicion in patients presenting with suggestive clinical features.
Collapse
Affiliation(s)
- David D. Nassoro
- Department of Internal MedicineMbeya Zonal Referral HospitalMbeyaTanzania
- Department of Internal MedicineThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Liset Torres
- Department of PathologyMbeya Zonal Referral HospitalMbeyaTanzania
- Department of PathologyThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Rehema Marando
- Department of Pediatrics and Child HealthMbeya Zonal Referral HospitalMbeyaTanzania
- Department of Pediatrics and Child HealthThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Lazaro Mboma
- Department of SurgeryMbeya Zonal Referral HospitalMbeyaTanzania
- Department of SurgeryThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Seraphine Mushi
- Department of PhysiotherapyMbeya Zonal Referral HospitalMbeyaTanzania
| | - Issakwisa Habakkuk Mwakyula
- Department of Internal MedicineMbeya Zonal Referral HospitalMbeyaTanzania
- Department of Internal MedicineThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| |
Collapse
|
28
|
"Betwixt Mine Eye and Heart a League Is Took": The Progress of Induced Pluripotent Stem-Cell-Based Models of Dystrophin-Associated Cardiomyopathy. Int J Mol Sci 2020; 21:ijms21196997. [PMID: 32977524 PMCID: PMC7582534 DOI: 10.3390/ijms21196997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/17/2020] [Accepted: 09/21/2020] [Indexed: 12/19/2022] Open
Abstract
The ultimate goal of precision disease modeling is to artificially recreate the disease of affected people in a highly controllable and adaptable external environment. This field has rapidly advanced which is evident from the application of patient-specific pluripotent stem-cell-derived precision therapies in numerous clinical trials aimed at a diverse set of diseases such as macular degeneration, heart disease, spinal cord injury, graft-versus-host disease, and muscular dystrophy. Despite the existence of semi-adequate treatments for tempering skeletal muscle degeneration in dystrophic patients, nonischemic cardiomyopathy remains one of the primary causes of death. Therefore, cardiovascular cells derived from muscular dystrophy patients' induced pluripotent stem cells are well suited to mimic dystrophin-associated cardiomyopathy and hold great promise for the development of future fully effective therapies. The purpose of this article is to convey the realities of employing precision disease models of dystrophin-associated cardiomyopathy. This is achieved by discussing, as suggested in the title echoing William Shakespeare's words, the settlements (or "leagues") made by researchers to manage the constraints ("betwixt mine eye and heart") distancing them from achieving a perfect precision disease model.
Collapse
|
29
|
Adipogenesis of skeletal muscle fibro/adipogenic progenitors is affected by the WNT5a/GSK3/β-catenin axis. Cell Death Differ 2020; 27:2921-2941. [PMID: 32382110 DOI: 10.1038/s41418-020-0551-y] [Citation(s) in RCA: 91] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/31/2022] Open
Abstract
Fibro/Adipogenic Progenitors (FAPs) are muscle-interstitial progenitors mediating pro-myogenic signals that are critical for muscle homeostasis and regeneration. In myopathies, the autocrine/paracrine constraints controlling FAP adipogenesis are released causing fat infiltrates. Here, by combining pharmacological screening, high-dimensional mass cytometry and in silico network modeling with the integration of single-cell/bulk RNA sequencing data, we highlighted the canonical WNT/GSK/β-catenin signaling as a crucial pathway modulating FAP adipogenesis triggered by insulin signaling. Consistently, pharmacological blockade of GSK3, by the LY2090314 inhibitor, stabilizes β-catenin and represses PPARγ expression abrogating FAP adipogenesis ex vivo while limiting fatty degeneration in vivo. Furthermore, GSK3 inhibition improves the FAP pro-myogenic role by efficiently stimulating, via follistatin secretion, muscle satellite cell (MuSC) differentiation into mature myotubes. Combining, publicly available single-cell RNAseq datasets, we characterize FAPs as the main source of WNT ligands inferring their potential in mediating autocrine/paracrine responses in the muscle niche. Lastly, we identify WNT5a, whose expression is impaired in dystrophic FAPs, as a crucial WNT ligand able to restrain the detrimental adipogenic differentiation drift of these cells through the positive modulation of the β-catenin signaling.
Collapse
|
30
|
Reggio A, Rosina M, Krahmer N, Palma A, Petrilli LL, Maiolatesi G, Massacci G, Salvatori I, Valle C, Testa S, Gargioli C, Fuoco C, Castagnoli L, Cesareni G, Sacco F. Metabolic reprogramming of fibro/adipogenic progenitors facilitates muscle regeneration. Life Sci Alliance 2020; 3:3/3/e202000646. [PMID: 32019766 PMCID: PMC7003708 DOI: 10.26508/lsa.202000660] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 01/28/2020] [Indexed: 12/25/2022] Open
Abstract
High-fat diet ameliorates muscle dystrophic phenotype by promoting the FAP-dependent myogenesis of satellite cells. In Duchenne muscular dystrophy (DMD), the absence of the dystrophin protein causes a variety of poorly understood secondary effects. Notably, muscle fibers of dystrophic individuals are characterized by mitochondrial dysfunctions, as revealed by a reduced ATP production rate and by defective oxidative phosphorylation. Here, we show that in a mouse model of DMD (mdx), fibro/adipogenic progenitors (FAPs) are characterized by a dysfunctional mitochondrial metabolism which correlates with increased adipogenic potential. Using high-sensitivity mass spectrometry–based proteomics, we report that a short-term high-fat diet (HFD) reprograms dystrophic FAP metabolism in vivo. By combining our proteomic dataset with a literature-derived signaling network, we revealed that HFD modulates the β-catenin–follistatin axis. These changes are accompanied by significant amelioration of the histological phenotype in dystrophic mice. Transplantation of purified FAPs from HFD-fed mice into the muscles of dystrophic recipients demonstrates that modulation of FAP metabolism can be functional to ameliorate the dystrophic phenotype. Our study supports metabolic reprogramming of muscle interstitial progenitor cells as a novel approach to alleviate some of the adverse outcomes of DMD.
Collapse
Affiliation(s)
- Alessio Reggio
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Marco Rosina
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Natalie Krahmer
- Department Proteomics and Signal Transduction, Max-Planck Institute of Biochemistry, Martinsried, Germany
| | - Alessandro Palma
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Giorgia Massacci
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Illari Salvatori
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Cristiana Valle
- Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy.,Institute of Translational Pharmacology, Consiglio Nazionale delle Ricerche (CNR), Rome, Italy
| | - Stefano Testa
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Cesare Gargioli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Claudia Fuoco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Luisa Castagnoli
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Gianni Cesareni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy .,Fondazione Santa Lucia Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Francesca Sacco
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| |
Collapse
|
31
|
Sitzia C, Meregalli M, Belicchi M, Farini A, Arosio M, Bestetti D, Villa C, Valenti L, Brambilla P, Torrente Y. Preliminary Evidences of Safety and Efficacy of Flavonoids- and Omega 3-Based Compound for Muscular Dystrophies Treatment: A Randomized Double-Blind Placebo Controlled Pilot Clinical Trial. Front Neurol 2019; 10:755. [PMID: 31396142 PMCID: PMC6664031 DOI: 10.3389/fneur.2019.00755] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
Background: Nutritional compounds can exert both anti-inflammatory and anti-oxidant effects. Since these events exacerbate the pathophysiology of muscular dystrophies, we investigated nutraceutical supplementation as an adjuvant therapy in dystrophic patients, to low costs and easy route of administration. Moreover, this treatment could represent an alternative therapeutic strategy for dystrophic patients who do not respond to corticosteroid treatment. Objective: A 24 weeks randomized double-blind placebo-controlled clinical study was aimed at evaluating the safety and efficacy of daily oral administration of flavonoids- and omega3-based natural supplement (FLAVOMEGA) in patients affected by muscular dystrophy with recognized muscle inflammation. Design: We screened 60 patients diagnosed for Duchenne (DMD), Facioscapulohumeral (FSHD), and Limb Girdle Muscular Dystrophy (LGMD). Using a computer-generated random allocation sequence, we stratified patients in a 2:1:1 ratio (DMD:FSHD:LGMD) to one of two treatment groups: continuous FLAVOMEGA, continuous placebo. Of 29 patients included, only 24 completed the study: 15 were given FLAVOMEGA, 14 placebo. Results: FLAVOMEGA was well tolerated with no reported adverse events. Significant treatment differences in the change from baseline in 6 min walk distance (6MWD; secondary efficacy endpoint) (P = 0.033) and in isokinetic knee extension (P = 0.039) (primary efficacy endpoint) were observed in LGMD and FSHD subjects. Serum CK levels (secondary efficacy endpoint) decreased in all FLAVOMEGA treated groups with significant difference in DMD subjects (P = 0.039). Conclusions: Although the small number of patients and the wide range of disease severity among patients reduced statistical significance, we obtained an optimal profile of safety and tolerability for the compound, showing valuable data of efficacy in primary and secondary endpoints. Trial registration number: NCT03317171 Retrospectively registered 25/10/2017
Collapse
Affiliation(s)
- Clementina Sitzia
- Stem Cell Laboratory, Unit of Neurology, Department of Pathophysiology and Transplantation, Centro Dino Ferrari, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Mirella Meregalli
- Stem Cell Laboratory, Unit of Neurology, Department of Pathophysiology and Transplantation, Centro Dino Ferrari, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marzia Belicchi
- Stem Cell Laboratory, Unit of Neurology, Department of Pathophysiology and Transplantation, Centro Dino Ferrari, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Andrea Farini
- Stem Cell Laboratory, Unit of Neurology, Department of Pathophysiology and Transplantation, Centro Dino Ferrari, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maddalena Arosio
- Service of Physiotherapy, San Raffaele Scientific Institute, Milan, Italy
| | - Denise Bestetti
- Bianchi Bonomi Haemophilia and Thrombosis Center, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Villa
- Stem Cell Laboratory, Unit of Neurology, Department of Pathophysiology and Transplantation, Centro Dino Ferrari, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Luca Valenti
- Department of Pathophysiology and Transplantation, Department of Transfusion Medicine and Hepatology, Translational Medicine, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda, Milan, Italy
| | - Paolo Brambilla
- Department of Laboratory Medicine, Desio Hospital, University Milano Bicocca, Milan, Italy
| | - Yvan Torrente
- Stem Cell Laboratory, Unit of Neurology, Department of Pathophysiology and Transplantation, Centro Dino Ferrari, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
32
|
Gallot YS, Straughn AR, Bohnert KR, Xiong G, Hindi SM, Kumar A. MyD88 is required for satellite cell-mediated myofiber regeneration in dystrophin-deficient mdx mice. Hum Mol Genet 2019; 27:3449-3463. [PMID: 30010933 DOI: 10.1093/hmg/ddy258] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Accepted: 07/09/2018] [Indexed: 02/07/2023] Open
Abstract
Duchenne muscular dystrophy (DMD), caused by mutations in the dystrophin gene, leads to severe muscle wasting and eventual death of the afflicted individuals, primarily due to respiratory failure. Deficit in myofiber regeneration, potentially due to an exhaustion of satellite cells, is one of the major pathological features of DMD. Myeloid differentiation primary response 88 (MyD88) is an adaptor protein that mediates activation of various inflammatory pathways in response to signaling from Toll-like receptors and interleukin-1 receptor. MyD88 also regulates cellular survival, proliferation and differentiation in a cell-autonomous manner. However, the role of MyD88 in satellite stem cell homeostasis and function in dystrophic muscle remains unknown. In this study, we demonstrate that tamoxifen-inducible deletion of MyD88 in satellite cells causes loss of skeletal muscle mass and strength in the mdx mouse model of DMD. Satellite cell-specific deletion of MyD88 inhibits myofiber regeneration and stimulates fibrogenesis in dystrophic muscle of mdx mice. Deletion of MyD88 also reduces the number of satellite cells and inhibits their fusion with injured myofibers in dystrophic muscle of mdx mice. Ablation of MyD88 in satellite cells increases the markers of M2 macrophages without having any significant effect on M1 macrophages and expression of inflammatory cytokines. Finally, we found that satellite cell-specific deletion of MyD88 leads to aberrant activation of Notch and Wnt signaling in skeletal muscle of mdx mice. Collectively, our results demonstrate that MyD88-mediated signaling in satellite cells is essential for the regeneration of injured myofibers in dystrophic muscle of mdx mice.
Collapse
Affiliation(s)
- Yann S Gallot
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Alex R Straughn
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Kyle R Bohnert
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Guangyan Xiong
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Sajedah M Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| |
Collapse
|
33
|
Santos NM, Rezende MDM, Terni A, Hayashi MCB, Fávero FM, Quadros AAJ, Reis LIOD, Adissi M, Langer AL, Fontes SV, Oliveira ASB. Perfil clínico e funcional dos pacientes com Distrofia Muscular de Duchenne assistidos na Associação Brasileira de Distrofia Muscular (ABDIM). ACTA ACUST UNITED AC 2019. [DOI: 10.34024/rnc.2006.v14.8782] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
A distrofia muscular de Duchenne (DMD) é uma doença hereditária progressiva, de herança recessiva ligada ao cromossomo X. As manifestações clínicas se iniciam na infância com enfraquecimento muscular progressivo. Objetivo: Foi caracterizar o perfil clínico e funcional apresentado pelos pacientes com DMD, que se encontravam em acompanhamento na Associação Brasileira de Distrofia Muscular (ABDIM). Método: Foram coletados os dados de 58 pacientes com o diagnóstico de DMD. Resultados: Os resultados obtidos caracterizaram os pacientes com DMD da ABDIM, proporcionando dados como: idade da população, complicações clínicas mais freqüentes e nível de independência em relação á funcionalidade, caracterizando a diversidade do perfil clínico e funcional desses pacientes. Conclusão: Concluiu-se que o perfil clínico e funcional dos pacientes da ABDIM apresentou-se heterogêneo, mostrando que a progressão da doença varia de sujeito para sujeito dentro da população estudada, mesmo levando em consideração a faixa etária.
Collapse
|
34
|
Lahouti AH, Christopher-Stine L. Inflammatory Muscle Diseases. Clin Immunol 2019. [DOI: 10.1016/b978-0-7020-6896-6.00056-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
35
|
Winters KV, Reynaud O, Novikov DS, Fieremans E, Kim SG. Quantifying myofiber integrity using diffusion MRI and random permeable barrier modeling in skeletal muscle growth and Duchenne muscular dystrophy model in mice. Magn Reson Med 2018; 80:2094-2108. [PMID: 29577406 PMCID: PMC6107391 DOI: 10.1002/mrm.27188] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 02/27/2018] [Accepted: 03/02/2018] [Indexed: 02/06/2023]
Abstract
PURPOSE To measure the microstructural changes during skeletal muscle growth and progressive pathologies using the random permeable model with diffusion MRI, and compare findings to conventional imaging modalities such as three-point Dixon and T2 imaging. METHODS In vivo and ex vivo DTI experiments with multiple diffusion times (20-700 ms) were completed on wild-type (n = 22) and muscle-dystrophic mdx mice (n = 8) at various developmental time points. The DTI data were analyzed with the random permeable model framework that provides estimates of the unrestricted diffusion coefficient (D0 ), membrane surface-to-volume ratio (S/V), and membrane permeability (κ). In addition, the MRI experiments included conventional measures, such as tissue fat fractions and T2 relaxation. RESULTS During normal muscle growth between week 4 and week 13, the in vivo S/V, fractional anisotropy, and fat fraction correlated positively with age (ρ = 0.638, 0.664, and 0.686, respectively), whereas T2 correlated negatively (ρ = -0.847). In mdx mice, all DTI random permeable model parameters and fat fraction had significant positive correlation with age, whereas fractional anisotropy and T2 did not have significant correlation with age. Histological measurements of the perimeter-to-area ratio served as a proxy for the model-derived S/V in the cylindrical myofiber geometry, and had a significant correlation with the ex vivo S/V (r = 0.71) as well as the in vivo S/V (r = 0.56). CONCLUSION The present study demonstrates that DTI at multiple diffusion times with the random permeable model analysis allows for noninvasively quantifying muscle fiber microstructural changes during both normal muscle growth and disease progression. Future studies can apply our technique to evaluate current and potential treatments to muscle myopathies.
Collapse
Affiliation(s)
- Kerryanne V. Winters
- Center for Advanced Imaging Innovation and Research (CAIR), New York, NY USA
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY USA
| | - Olivier Reynaud
- Center for Advanced Imaging Innovation and Research (CAIR), New York, NY USA
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY USA
| | - Dmitry S. Novikov
- Center for Advanced Imaging Innovation and Research (CAIR), New York, NY USA
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY USA
| | - Els Fieremans
- Center for Advanced Imaging Innovation and Research (CAIR), New York, NY USA
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY USA
| | - Sungheon Gene Kim
- Center for Advanced Imaging Innovation and Research (CAIR), New York, NY USA
- Bernard and Irene Schwartz Center for Biomedical Imaging, Department of Radiology, New York University School of Medicine, New York, NY USA
| |
Collapse
|
36
|
Banfi S, D'Antona G, Ruocco C, Meregalli M, Belicchi M, Bella P, Erratico S, Donato E, Rossi F, Bifari F, Lonati C, Campaner S, Nisoli E, Torrente Y. Supplementation with a selective amino acid formula ameliorates muscular dystrophy in mdx mice. Sci Rep 2018; 8:14659. [PMID: 30279586 PMCID: PMC6168581 DOI: 10.1038/s41598-018-32613-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 09/10/2018] [Indexed: 11/19/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is one of the most common and severe forms of muscular dystrophy. Oxidative myofibre content, muscle vasculature architecture and exercise tolerance are impaired in DMD. Several studies have demonstrated that nutrient supplements ameliorate dystrophic features, thereby enhancing muscle performance. Here, we report that dietary supplementation with a specific branched-chain amino acid-enriched mixture (BCAAem) increased the abundance of oxidative muscle fibres associated with increased muscle endurance in dystrophic mdx mice. Amelioration of the fatigue index in BCAAem-treated mdx mice was caused by a cascade of events in the muscle tissue, which were promoted by endothelial nitric oxide synthase (eNOS) activation and vascular endothelial growth factor (VEGF) expression. VEGF induction led to recruitment of bone marrow (BM)-derived endothelial progenitors (EPs), which increased the capillary density of dystrophic skeletal muscle. Functionally, BCAAem mitigated the dystrophic phenotype of mdx mice without inducing dystrophin protein expression or replacing the dystrophin-associated glycoprotein (DAG) complex in the membrane, which is typically lost in DMD. BCAAem supplementation could be an effective adjuvant strategy in DMD treatment.
Collapse
Affiliation(s)
- Stefania Banfi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122, Milan, Italy
| | - Giuseppe D'Antona
- Department of Public Health, Molecular and Forensic Medicine, and Sport Medicine Centre Voghera, University of Pavia, Pavia, 27100, Italy
| | - Chiara Ruocco
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, 20129, Italy
| | - Mirella Meregalli
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122, Milan, Italy
| | - Marzia Belicchi
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122, Milan, Italy
| | - Pamela Bella
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122, Milan, Italy
| | | | - Elisa Donato
- Centre for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia, Milan, 20139, Italy.,Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum, Heidelberg, Germany.,Heidelberg Institute for Stem Cell Technology and Experimental Medicine, Heidelberg, Germany
| | - Fabio Rossi
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, 20129, Italy
| | - Francesco Bifari
- Laboratory of Cell Metabolism and Regenerative Medicine, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, 20129, Milan, Italy
| | - Caterina Lonati
- Center for Surgical Research, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, 20122, Italy
| | - Stefano Campaner
- Centre for Genomic Science of IIT@SEMM, Istituto Italiano di Tecnologia, Milan, 20139, Italy
| | - Enzo Nisoli
- Center for Study and Research on Obesity, Department of Medical Biotechnology and Translational Medicine, University of Milan, Milan, 20129, Italy.
| | - Yvan Torrente
- Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, 20122, Milan, Italy.
| |
Collapse
|
37
|
Law ML, Prins KW, Olander ME, Metzger JM. Exacerbation of dystrophic cardiomyopathy by phospholamban deficiency mediated chronically increased cardiac Ca 2+ cycling in vivo. Am J Physiol Heart Circ Physiol 2018; 315:H1544-H1552. [PMID: 30118340 DOI: 10.1152/ajpheart.00341.2018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cardiomyopathy is a significant contributor to morbidity and mortality in Duchenne muscular dystrophy (DMD). Membrane instability, leading to intracellular Ca2+ mishandling and overload, causes myocyte death and subsequent fibrosis in DMD cardiomyopathy. On a cellular level, cardiac myocytes from mdx mice have dysregulated Ca2+ handling, including increased resting Ca2+ and slow Ca2+ decay, especially evident under stress conditions. Sarco(endo)plasmic reticulum Ca2+ ATPase and its regulatory protein phospholamban (PLN) are potential therapeutic targets for DMD cardiomyopathy owing to their key role in regulating intracellular Ca2+ cycling. We tested the hypothesis that enhanced cardiac Ca2+ cycling would remediate cardiomyopathy caused by dystrophin deficiency. We used a genetic complementation model approach by crossing dystrophin-deficient mdx mice with PLN knockout (PLNKO) mice [termed double-knockout (DKO) mice]. As expected, adult cardiac myocytes isolated from DKO mice exhibited increased contractility and faster relaxation associated with increased Ca2+ transient peak height and faster Ca2+ decay rate compared with control mice. However, compared with wild-type, mdx, and PLNKO mice, DKO mice unexpectedly had reduced in vivo systolic and diastolic function as measured by echocardiography. Furthermore, Evans blue dye uptake was increased in DKO hearts compared with control, mdx, and PLNKO hearts, demonstrating increased membrane damage, which subsequently led to increased fibrosis in the DKO myocardium in vivo. In conclusion, despite enhanced intracellular Ca2+ handling at the myocyte level, DMD cardiomyopathy was exacerbated owing to unregulated chronic increases in Ca2+ cycling in DKO mice in vivo. These findings have potentially important implications for ongoing therapeutic strategies for the dystrophic heart. NEW & NOTEWORTHY This study examined the effects of phospholamban ablation on the pathophysiology of cardiomyopathy in dystrophin-deficient mice. In this setting, contractility and Ca2+ cycling were enhanced in isolated myocytes; however, in vivo heart function was diminished. Additionally, sarcolemmal integrity was compromised and fibrosis was increased. This is the first study, to our knowledge, examining unregulated Ca2+ cycling in the dystrophin-deficient heart. Results from this study have implications for potential therapies targeting Ca2+ handling in dystrophic cardiomyopathy. Listen to this article's corresponding podcast at https://ajpheart.podbean.com/e/unregulated-ca2-cycling-exacerbates-dmd-cardiomyopathy/ .
Collapse
Affiliation(s)
- Michelle L Law
- Department of Integrative Biology and Physiology, University of Minnesota Medical School , Minneapolis, Minnesota
| | - Kurt W Prins
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical School , Minneapolis, Minnesota
| | - Megan E Olander
- Department of Integrative Biology and Physiology, University of Minnesota Medical School , Minneapolis, Minnesota
| | - Joseph M Metzger
- Department of Integrative Biology and Physiology, University of Minnesota Medical School , Minneapolis, Minnesota
| |
Collapse
|
38
|
Do TN, Street N, Donnelly J, Adams MM, Cunniff C, Fox DJ, Weinert RO, Oleszek J, Romitti PA, Westfield CP, Bolen J. Muscular Dystrophy Surveillance, Tracking, and Research Network pilot: Population-based surveillance of major muscular dystrophies at four U.S. sites, 2007-2011. Birth Defects Res 2018; 110:1404-1411. [PMID: 30070776 DOI: 10.1002/bdr2.1371] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Revised: 06/15/2018] [Accepted: 06/21/2018] [Indexed: 11/10/2022]
Abstract
BACKGROUND For 10 years, the Muscular Dystrophy Surveillance, Tracking, and Research Network (MD STARnet) conducted surveillance for Duchenne and Becker muscular dystrophy (DBMD). We piloted expanding surveillance to other MDs that vary in severity, onset, and sources of care. METHODS Our retrospective surveillance included individuals diagnosed with one of nine eligible MDs before or during the study period (January 2007-December 2011), one or more health encounters, and residence in one of four U.S. sites (Arizona, Colorado, Iowa, or western New York) at any time within the study period. We developed case definitions, surveillance protocols, and software applications for medical record abstraction, clinical review, and data pooling. Potential cases were identified by International Classification of Diseases, Ninth Revision, Clinical Modification (ICD-9-CM) codes 359.0, 359.1, and 359.21 and International Classification of Diseases, Tenth Revision (ICD-10) codes G71.0 and G71.1. Descriptive statistics were compared by MD type. Percentage of MD cases identified by each ICD-9-CM code was calculated. RESULTS Of 2,862 cases, 32.9% were myotonic, dystrophy 25.8% DBMD, 9.7% facioscapulohumeral MD, and 9.1% limb-girdle MD. Most cases were male (63.6%), non-Hispanic (59.8%), and White (80.2%). About, half of cases were genetically diagnosed in self (39.1%) or family (6.2%). About, half had a family history of MD (48.9%). The hereditary progressive MD code (359.1) was the most common code for identifying eligible cases. The myotonic code (359.21) identified 83.4% of eligible myotonic dystrophy cases (786/943). CONCLUSIONS MD STARnet is the only multisite, population-based active surveillance system available for MD in the United States. Continuing our expanded surveillance will contribute important epidemiologic and health outcome information about several MDs.
Collapse
Affiliation(s)
- ThuyQuynh N Do
- Centers for Disease Control and Prevention, National Center on Birth Defects and Developmental Disabilities, Atlanta, Georgia
| | - Natalie Street
- Centers for Disease Control and Prevention, National Center on Birth Defects and Developmental Disabilities, Atlanta, Georgia
| | - Jennifer Donnelly
- Colorado Department of Public Health & Environment, Denver, Colorado
| | | | | | - Deborah J Fox
- Bureau of Environmental and Occupational Epidemiology, New York State Department of Health, Albany, New York
| | - Richard O Weinert
- Colorado Department of Public Health & Environment, Denver, Colorado
| | - Joyce Oleszek
- University of Colorado, Denver and Children's Hospital, Aurora, Colorado
| | - Paul A Romitti
- Department of Epidemiology, College of Public Health, University of Iowa, Iowa City, Iowa
| | - Christina P Westfield
- Bureau of Environmental and Occupational Epidemiology, New York State Department of Health, Albany, New York
| | - Julie Bolen
- Centers for Disease Control and Prevention, National Center on Birth Defects and Developmental Disabilities, Atlanta, Georgia
| | | |
Collapse
|
39
|
Deepha S, Vengalil S, Preethish-Kumar V, Polavarapu K, Nalini A, Gayathri N, Purushottam M. MLPA identification of dystrophin mutations and in silico evaluation of the predicted protein in dystrophinopathy cases from India. BMC MEDICAL GENETICS 2017; 18:67. [PMID: 28610567 PMCID: PMC5470271 DOI: 10.1186/s12881-017-0431-6] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/31/2017] [Indexed: 11/18/2022]
Abstract
Background Duchenne muscular dystrophy (DMD) and Becker muscular dystrophy (BMD) are X-linked recessive disorders caused by mutations in the DMD gene. The aim of this study was to predict the effect of gene mutations on the dystrophin protein and study its impact on clinical phenotype. Methods In this study, 415 clinically diagnosed patients were tested for mutations by Multiplex ligation dependent probe amplification (MLPA). Muscle biopsy was performed in 34 patients with negative MLPA. Phenotype-genotype correlation was done using PROVEAN, hydrophobicity and eDystrophin analysis. We have utilized bioinformatics tools in order to evaluate the observed mutations both at the level of primary as well as secondary structure. Results Mutations were identified in 75.42% cases, of which there were deletions in 91.6% and duplications in 8.30%. As per the reading frame rule, 84.6% out-of frame and 15.3% in-frame mutations were noted. Exon 50 was the most frequently deleted exon and the exon 45–52 region was the hot-spot for deletions in this cohort. There was no correlation noted between age of onset or creatine kinase (CK) values with extent of gene mutation. The PROVEAN analysis showed a deleterious effect in 94.5% cases and a neutral effect in 5.09% cases. Mutations in exon 45–54 (out of frame) and exon 46–54 (in-frame) regions in the central rod domain of dystrophin showed more negative scores compared to other domains in the present study. Hydrophobicity profile analysis showed that the hydrophobic regions I & III were equally affected. Analysis of deletions in hinge III hydrophobic region by the eDystrophin programme also predicted a hybrid repeat seen to be associated with a BMD like disease progression, thus making the hinge III region relatively tolerant to mutations. Conclusions We found that, while the predictions made by the software utilized might have overall significance, the results were not convincing on a case by case basis. This reflects the inadequacy of the currently available tools and also underlines the possible inadequacy of MLPA to detect other minor mutations that might enhance or suppress the effect of the primary mutation in this large gene. Next Generation Sequencing or targeted Sanger sequencing on a case by case basis might improve phenotype- genotype correlation. Electronic supplementary material The online version of this article (doi:10.1186/s12881-017-0431-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sekar Deepha
- Department of Neuropathology, Neuromuscular Laboratory, Bengaluru, Karnataka, India
| | - Seena Vengalil
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Veeramani Preethish-Kumar
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Kiran Polavarapu
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Atchayaram Nalini
- Department of Neurology, National Institute of Mental Health and Neurosciences, Bengaluru, Karnataka, India
| | - Narayanappa Gayathri
- Department of Neuropathology, Neuromuscular Laboratory, Bengaluru, Karnataka, India
| | - Meera Purushottam
- Molecular Genetics Laboratory, Neurobiology Research Centre, Department of Psychiatry, National Institute of Mental Health and Neurosciences, Hosur Road, Bangalore, 560029, Karnataka, India.
| |
Collapse
|
40
|
Bell JM, Shields MD, Watters J, Hamilton A, Beringer T, Elliott M, Quinlivan R, Tirupathi S, Blackwood B. Interventions to prevent and treat corticosteroid-induced osteoporosis and prevent osteoporotic fractures in Duchenne muscular dystrophy. Cochrane Database Syst Rev 2017; 1:CD010899. [PMID: 28117876 PMCID: PMC6464928 DOI: 10.1002/14651858.cd010899.pub2] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Corticosteroid treatment is considered the 'gold standard' for Duchenne muscular dystrophy (DMD); however, it is also known to induce osteoporosis and thus increase the risk of vertebral fragility fractures. Good practice in the care of those with DMD requires prevention of these adverse effects. Treatments to increase bone mineral density include bisphosphonates and vitamin D and calcium supplements, and in adolescents with pubertal delay, testosterone. Bone health management is an important part of lifelong care for patients with DMD. OBJECTIVES To assess the effects of interventions to prevent or treat osteoporosis in children and adults with DMD taking long-term corticosteroids; to assess the effects of these interventions on the frequency of vertebral fragility fractures and long-bone fractures, and on quality of life; and to assess adverse events. SEARCH METHODS On 12 September 2016, we searched the Cochrane Neuromuscular Specialised Register, CENTRAL, MEDLINE, Embase, and CINAHL Plus to identify potentially eligible trials. We also searched the Web of Science ISI Proceedings (2001 to September 2016) and three clinical trials registries to identify unpublished studies and ongoing trials. We contacted correspondence authors of the included studies in the review to obtain information on unpublished studies or work in progress. SELECTION CRITERIA We considered for inclusion in the review randomised controlled trials (RCTs) and quasi-RCTs involving any bone health intervention for corticosteroid-induced osteoporosis and fragility fractures in children, adolescents, and adults with a confirmed diagnosis of DMD. The interventions might have included oral and intravenous bisphosphonates, vitamin D supplements, calcium supplements, dietary calcium, testosterone, and weight-bearing activity. DATA COLLECTION AND ANALYSIS Two review authors independently assessed reports and selected potential studies for inclusion, following standard Cochrane methodology. We contacted study authors to obtain further information for clarification on published work, unpublished studies, and work in progress. MAIN RESULTS We identified 18 potential studies, of which two, currently reported only as abstracts, met the inclusion criteria for this review. Too little information was available for us to present full results or adequately assess risk of bias. The participants were children aged five to 15 years with DMD, ambulant and non-ambulant. The interventions were risedronate versus no treatment in one trial (13 participants) and whole-body vibration versus a placebo device in the second (21 participants). Both studies reported improved bone mineral density with the active treatments, with no improvement in the control groups, but the abstracts did not compare treatment and control conditions. All children tolerated whole-body vibration treatment. No study provided information on adverse events. Two studies are ongoing: one investigating whole-body vibration, the other investigating zoledronic acid. AUTHORS' CONCLUSIONS We know of no high-quality evidence from RCTs to guide use of treatments to prevent or treat corticosteroid-induced osteoporosis and reduce the risk of fragility fractures in children and adults with DMD; only limited results from two trials reported in abstracts were available. We await formal trial reports. Findings from two ongoing relevant studies and two trials, for which only abstracts are available, will be important in future updates of this review.
Collapse
Affiliation(s)
- Jennifer M Bell
- Queen's University BelfastCentre for Experimental Medicine, School of Medicine, Dentistry and Biomedical SciencesRoom 02.041, 2nd FloorMulhouse, Grosvenor RoadBelfastNorthern IrelandUKBT12 6BJ
| | - Michael D Shields
- Queen's University BelfastCentre for Experimental Medicine, School of Medicine, Dentistry and Biomedical SciencesRoom 02.041, 2nd FloorMulhouse, Grosvenor RoadBelfastNorthern IrelandUKBT12 6BJ
| | - Janet Watters
- Belfast Health and Social Care TrustGP Out of Hours ServiceBelfastNorthern IrelandUK
| | - Alistair Hamilton
- Belfast Health and Social Care TrustWithers Orthopaedic CentreMusgrave Park Hospital, Royal Group of Hospitals,Stockman's LaneBelfastNorthern IrelandUK
| | - Timothy Beringer
- Belfast Health and Social Care TrustDepartment of Care for the ElderyFlorence Elliot CentreRoyal Victoria HospitalBelfastNorthern IrelandUKBT12 6BA
| | - Mark Elliott
- Musgrave Park Hospital, Belfast Health and Social Care TrustBelfastUK
| | - Rosaline Quinlivan
- UCL Institute of Neurology and National Hospital for Neurology and Neurosurgery and Great Ormond StreetMRC Centre for Neuromuscular Diseases and Dubowitz Neuromuscular CentrePO Box 114LondonUKWC1B 3BN
| | - Sandya Tirupathi
- Royal Belfast Hospital for Sick ChildrenPaediatric Neurology180 Falls RoadBelfastUKBT12 6BE
| | - Bronagh Blackwood
- Queen's University BelfastCentre for Experimental Medicine, School of Medicine, Dentistry and Biomedical SciencesRoom 02.041, 2nd FloorMulhouse, Grosvenor RoadBelfastNorthern IrelandUKBT12 6BJ
| | | |
Collapse
|
41
|
Iyer SR, Xu S, Stains JP, Bennett CH, Lovering RM. Superparamagnetic Iron Oxide Nanoparticles in Musculoskeletal Biology. TISSUE ENGINEERING PART B-REVIEWS 2017; 23:373-385. [PMID: 27998240 DOI: 10.1089/ten.teb.2016.0437] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The use of platelet-rich plasma and mesenchymal stem cells has garnered much attention in orthopedic medicine, focusing on the biological aspects of cell function. However, shortly after systemic delivery, or even a local injection, few of the transplanted stem cells or platelets remain at the target site. Improvement in delivery, and the ability to track and monitor injected cells, would greatly improve clinical translation. Nanoparticles can effectively and quickly label most cells in vitro, and evidence to date suggests such labeling does not compromise the proliferation or differentiation of cells. A specific type of nanoparticle, the superparamagnetic iron oxide nanoparticle (SPION), is already employed as a magnetic resonance imaging (MRI) contrast agent. SPIONs can be coupled with cells or bioactive molecules (antibodies, proteins, drugs, etc.) to form an injectable complex for in vivo use. The biocompatibility, magnetic properties, small size, and custom-made surface coatings also enable SPIONs to be used for delivering and monitoring of small molecules, drugs, and cells, specifically to muscle, bone, or cartilage. Because SPIONs consist of cores made of iron oxides, targeting of SPIONs to a specific muscle, bone, or joint in the body can be enhanced with the help of applied gradient magnetic fields. Moreover, MRI has a high sensitivity to SPIONs and can be used for noninvasive determination of successful delivery and monitoring distribution in vivo. Gaps remain in understanding how the physical and chemical properties of nanomaterials affect biological systems. Nonetheless, SPIONs hold great promise for regenerative medicine, and progress is being made rapidly toward clinical applications in orthopedic medicine.
Collapse
Affiliation(s)
- Shama R Iyer
- 1 Department of Orthopaedics, University of Maryland School of Medicine , Baltimore, Maryland
| | - Su Xu
- 2 Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine , Baltimore, Maryland
| | - Joseph P Stains
- 1 Department of Orthopaedics, University of Maryland School of Medicine , Baltimore, Maryland
| | - Craig H Bennett
- 1 Department of Orthopaedics, University of Maryland School of Medicine , Baltimore, Maryland
| | - Richard M Lovering
- 1 Department of Orthopaedics, University of Maryland School of Medicine , Baltimore, Maryland.,3 Department of Physiology, University of Maryland School of Medicine , Baltimore, Maryland
| |
Collapse
|
42
|
Bidiwala A, Volpe L, Halaby C, Fazzari M, Valsamis C, Pirzada M. A comparison of high frequency chest wall oscillation and intrapulmonary percussive ventilation for airway clearance in pediatric patients with tracheostomy. Postgrad Med 2016; 129:276-282. [DOI: 10.1080/00325481.2017.1264854] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Aneela Bidiwala
- Division of Respiratory and Sleep Medicine, Children’s Hospital at Montefiore, Bronx, NY, USA
| | - Linda Volpe
- St Mary’s Health Care System for Children, Bayside, NY, USA
| | - Claudia Halaby
- Pediatric Pulmonary Division, Winthrop University Hospital, Mineola, NY, USA
| | - Melissa Fazzari
- Biostatic Department, Winthrop University Hospital, Mineola, NY, USA
| | - Christina Valsamis
- Pediatric Pulmonary Division, Winthrop University Hospital, Mineola, NY, USA
| | - Melodi Pirzada
- Pediatric Pulmonary Division, Winthrop University Hospital, Mineola, NY, USA
| |
Collapse
|
43
|
Farini A, Sitzia C, Cassani B, Cassinelli L, Rigoni R, Colleoni F, Fusco N, Gatti S, Bella P, Villa C, Napolitano F, Maiavacca R, Bosari S, Villa A, Torrente Y. Therapeutic Potential of Immunoproteasome Inhibition in Duchenne Muscular Dystrophy. Mol Ther 2016; 24:1898-1912. [PMID: 27506451 DOI: 10.1038/mt.2016.162] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 07/28/2016] [Indexed: 01/24/2023] Open
Abstract
Duchenne muscular dystrophy is an inherited fatal genetic disease characterized by mutations in dystrophin gene, causing membrane fragility leading to myofiber necrosis and inflammatory cell recruitment in dystrophic muscles. The resulting environment enriched in proinflammatory cytokines, like IFN-γ and TNF-α, determines the transformation of myofiber constitutive proteasome into the immunoproteasome, a multisubunit complex involved in the activation of cell-mediate immunity. This event has a fundamental role in producing peptides for antigen presentation by MHC class I, for the immune response and also for cytokine production and T-cell differentiation. Here, we characterized for the first time the presence of T-lymphocytes activated against revertant dystrophin epitopes, in the animal model of Duchenne muscular dystrophy, the mdx mice. Moreover, we specifically blocked i-proteasome subunit LMP7, which was up-regulated in dystrophic skeletal muscles, and we demonstrated the rescue of the dystrophin expression and the amelioration of the dystrophic phenotype. The i-proteasome blocking lowered myofiber MHC class I expression and self-antigen presentation to T cells, thus reducing the specific antidystrophin T cell response, the muscular cell infiltrate, and proinflammatory cytokine production, together with muscle force recovery. We suggest that i-proteasome inhibition should be considered as new promising therapeutic approach for Duchenne muscular dystrophy pathology.
Collapse
Affiliation(s)
- Andrea Farini
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Clementina Sitzia
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy; Department of Laboratory Medicine, University Milano Bicocca, Desio Hospital, Desio (MB), Italy
| | - Barbara Cassani
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy; Humanitas Clinical and Research Center, Milan, Italy
| | - Letizia Cassinelli
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Rosita Rigoni
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy; Humanitas Clinical and Research Center, Milan, Italy
| | - Federica Colleoni
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Nicola Fusco
- Department of Pathophysiology and Organ Transplantation, University of Milan, Milan, Italy; Division of Pathology, Fondazione IRCCS Cà Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Stefano Gatti
- Center for Surgical Research, Fondazione IRCCS Cà Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Pamela Bella
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Chiara Villa
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy
| | - Filomena Napolitano
- Laboratorio di Chimica Clinica e Microbiologia, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Rita Maiavacca
- Laboratorio di Chimica Clinica e Microbiologia, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Italy
| | - Silvano Bosari
- Department of Pathophysiology and Organ Transplantation, University of Milan, Milan, Italy; Division of Pathology, Fondazione IRCCS Cà Granda - Ospedale Maggiore Policlinico, Milan, Italy
| | - Anna Villa
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy; Humanitas Clinical and Research Center, Milan, Italy
| | - Yvan Torrente
- Stem Cell Laboratory, Department of Pathophysiology and Transplantation, Università degli Studi di Milano, Unit of Neurology, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Centro Dino Ferrari, Milan, Italy.
| |
Collapse
|
44
|
Noncoding RNAs in the regulation of skeletal muscle biology in health and disease. J Mol Med (Berl) 2016; 94:853-66. [PMID: 27377406 DOI: 10.1007/s00109-016-1443-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Revised: 05/11/2016] [Accepted: 06/20/2016] [Indexed: 12/22/2022]
Abstract
Skeletal muscle is composed of multinucleated myofibers that arise from the fusion of myoblasts during development. Skeletal muscle is essential for various body functions such as maintaining posture, locomotion, breathing, and metabolism. Skeletal muscle undergoes remarkable adaptations in response to environmental stimuli leading to atrophy or hypertrophy. Moreover, degeneration of skeletal muscle is a common feature in a number of muscular disorders including muscular dystrophy. Emerging evidence suggests that noncoding RNAs, such as microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), are critical for skeletal muscle physiology. Several miRNAs and lncRNAs have now been found to control skeletal muscle development and regeneration. Noncoding RNAs also play an important role in the regulation of skeletal muscle mass in adults. Furthermore, aberrant expression of miRNAs and lncRNAs has been observed in several muscular disorders. In this article, we discuss the mechanisms of action of miRNAs and lncRNAs in skeletal muscle formation, growth, regeneration, and disease. We further highlight potential therapeutic strategies for utilizing noncoding RNAs to improve skeletal muscle function.
Collapse
|
45
|
Abstract
Wnt signaling plays key roles in many aspects of development. In this review, we will briefly describe the components of signaling pathways induced by Wnt ligands and then describe the current state of research as this applies to aspects of development and disease as it relates to skeletal muscle and bone. We will conclude with a discussion of the parallels and differences in Wnt signaling in these two contexts and how these pathways are being (or could potentially be) targeted for therapeutic treatment of musculoskeletal diseases. This article is part of a Special Issue entitled "Muscle Bone Interactions".
Collapse
Affiliation(s)
- Michael A Rudnicki
- Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Ontario, Canada; Department Medicine, University of Ottawa, Ottawa, Ontario, Canada.
| | - Bart O Williams
- Center for Skeletal Disease and Tumor Metastasis, Van Andel Research Institute, Grand Rapids, MI, USA.
| |
Collapse
|
46
|
Genetic and clinical profile of patients of Duchenne muscular dystrophy: Experience from a tertiary care center in Eastern India. Indian Pediatr 2015; 52:481-4. [DOI: 10.1007/s13312-015-0660-8] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
47
|
Comim CM, Mathia GB, Hoepers A, Tuon L, Kapczinski F, Dal-Pizzol F, Quevedo J, Rosa MI. Neurotrophins, cytokines, oxidative parameters and funcionality in Progressive Muscular Dystrophies. AN ACAD BRAS CIENC 2015; 87:1809-18. [PMID: 25910175 DOI: 10.1590/0001-3765201520140508] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
We investigated the levels of brain derived-neurotrophic factor (BDNF), cytokines and oxidative parameters in serum and tried to correlate them with the age and functionality of patients with Progressive Muscle Dystrophies (PMD). The patients were separated into six groups (case and controls pared by age and gender), as follows: Duchenne Muscular Dystrophy (DMD); Steinert Myotonic Dystrophy (SMD); and Limb-girdle Muscular Dystrophy type-2A (LGMD2A). DMD patients (± 17.9 years old) had a decrease of functionality, an increase in the IL-1β and TNF-α levels and a decrease of IL-10 levels and superoxide dismutase activity in serum. SMD patients (± 25.8 years old) had a decrease of BDNF and IL-10 levels and superoxide dismutase activity and an increase of IL-1β levels in serum. LGMD2A patients (± 27.7 years old) had an decrease only in serum levels of IL-10. This research showed the first evidence of BDNF involvement in the SMD patients and a possible unbalance between pro-inflammatory and anti-inflammatory cytokine levels, along with decreased superoxide dismutase activity in serum of DMD and SMD patients.
Collapse
Affiliation(s)
| | | | | | - Lisiane Tuon
- Universidade do Extremo Sul Catarinense, Criciúma, SC, BR
| | - Flávio Kapczinski
- Centro de Pesquisas, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, BR
| | | | - João Quevedo
- Universidade do Extremo Sul Catarinense, Criciúma, SC, BR
| | - Maria I Rosa
- Universidade do Extremo Sul Catarinense, Criciúma, SC, BR
| |
Collapse
|
48
|
Santacatterina F, Chamorro M, de Arenas CN, Navarro C, Martín MA, Cuezva JM, Sánchez-Aragó M. Quantitative analysis of proteins of metabolism by reverse phase protein microarrays identifies potential biomarkers of rare neuromuscular diseases. J Transl Med 2015; 13:65. [PMID: 25880557 PMCID: PMC4342896 DOI: 10.1186/s12967-015-0424-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Accepted: 01/30/2015] [Indexed: 11/16/2022] Open
Abstract
Background Muscle diseases have been associated with changes in the expression of proteins involved in energy metabolism. To this aim we have developed a number of monoclonal antibodies against proteins of energy metabolism. Methods Herein, we have used Reverse Phase Protein Microarrays (RPMA), a high throughput technique, to investigate quantitative changes in protein expression with the aim of identifying potential biomarkers in rare neuromuscular diseases. A cohort of 73 muscle biopsies that included samples from patients diagnosed of Duchenne (DMD), Becker (BMD), symptomatic forms of DMD and BMD in female carriers (Xp21 Carriers), Limb Girdle Muscular Dystrophy Type 2C (LGMD2C), neuronal ceroid lipofuscinosis (NCL), glycogenosis type V (Mc Ardle disease), isolated mitochondrial complex I deficiency, intensive care unit myopathy and control donors were investigated. The nineteen proteins of energy metabolism studied included members of the mitochondrial oxidation of pyruvate, the tricarboxylic acid cycle, β-oxidation of fatty acids, electron transport and oxidative phosphorylation, glycogen metabolism, glycolysis and oxidative stress using highly specific antibodies. Results The results indicate that the phenotype of energy metabolism offers potential biomarkers that could be implemented to refine the understanding of the biological principles of rare diseases and, eventually, the management of these patients. Conclusions We suggest that some biomarkers of energy metabolism could be translated into the clinics to contribute to the improvement of the clinical handling of patients affected by rare diseases. Electronic supplementary material The online version of this article (doi:10.1186/s12967-015-0424-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Fulvio Santacatterina
- Departamento de Biología Molecular, Centro de Biología Molecular, c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain.
| | - Margarita Chamorro
- Departamento de Biología Molecular, Centro de Biología Molecular, c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain.
| | - Cristina Núñez de Arenas
- Departamento de Biología Molecular, Centro de Biología Molecular, c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain.
| | - Carmen Navarro
- Instituto de Investigación Biomédico de Vigo (IBIV), Hospital Universitario de Vigo, Meixoeiro, 36200, Vigo, Spain.
| | - Miguel A Martín
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain. .,Laboratorio de Enfermedades Mitocondriales y Neuromusculares, Hospital Universitario 12 de Octubre, 28041, Madrid, Spain.
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular, c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain.
| | - María Sánchez-Aragó
- Departamento de Biología Molecular, Centro de Biología Molecular, c/ Nicolás Cabrera 1, Universidad Autónoma de Madrid, 28049, Madrid, Spain. .,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain. .,Instituto de Investigación Hospital 12 de Octubre, ISCIII, Madrid, Spain.
| |
Collapse
|
49
|
Influence of immune responses in gene/stem cell therapies for muscular dystrophies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:818107. [PMID: 24959590 PMCID: PMC4052166 DOI: 10.1155/2014/818107] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 04/07/2014] [Accepted: 04/30/2014] [Indexed: 02/06/2023]
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of diseases, caused by mutations in different components of sarcolemma, extracellular matrix, or enzymes. Inflammation and innate or adaptive immune response activation are prominent features of MDs. Various therapies under development are directed toward rescuing the dystrophic muscle damage using gene transfer or cell therapy. Here we discussed current knowledge about involvement of immune system responses to experimental therapies in MDs.
Collapse
|
50
|
Kayman-Kurekci G, Talim B, Korkusuz P, Sayar N, Sarioglu T, Oncel I, Sharafi P, Gundesli H, Balci-Hayta B, Purali N, Serdaroglu-Oflazer P, Topaloglu H, Dincer P. Mutation in TOR1AIP1 encoding LAP1B in a form of muscular dystrophy: a novel gene related to nuclear envelopathies. Neuromuscul Disord 2014; 24:624-33. [PMID: 24856141 DOI: 10.1016/j.nmd.2014.04.007] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2014] [Revised: 04/21/2014] [Accepted: 04/24/2014] [Indexed: 11/26/2022]
Abstract
We performed genome-wide homozygosity mapping and mapped a novel myopathic phenotype to chromosomal region 1q25 in a consanguineous family with three affected individuals manifesting proximal and distal weakness and atrophy, rigid spine and contractures of the proximal and distal interphalangeal hand joints. Additionally, cardiomyopathy and respiratory involvement were noted. DNA sequencing of torsinA-interacting protein 1 (TOR1AIP1) gene encoding lamina-associated polypeptide 1B (LAP1B), showed a homozygous c.186delG mutation that causes a frameshift resulting in a premature stop codon (p.E62fsTer25). We observed that expression of LAP1B was absent in the patient skeletal muscle fibres. Ultrastructural examination showed intact sarcomeric organization but alterations of the nuclear envelope including nuclear fragmentation, chromatin bleb formation and naked chromatin. LAP1B is a type-2 integral membrane protein localized in the inner nuclear membrane that binds to both A- and B-type lamins, and is involved in the regulation of torsinA ATPase. Interestingly, luminal domain-like LAP1 (LULL1)-an endoplasmic reticulum-localized partner of torsinA-was overexpressed in the patient's muscle in the absence of LAP1B. Therefore, the findings suggest that LAP1 and LULL1 might have a compensatory effect on each other. This study expands the spectrum of genes associated with nuclear envelopathies and highlights the critical function for LAP1B in striated muscle.
Collapse
Affiliation(s)
- Gulsum Kayman-Kurekci
- Department of Medical Biology, Hacettepe University, Faculty of Medicine, Sihhiye, 06100 Ankara, Turkey
| | - Beril Talim
- Department of Pediatrics, Pathology Unit, Hacettepe University, Faculty of Medicine, Sihhiye, 06100 Ankara, Turkey
| | - Petek Korkusuz
- Department of Histology and Embryology, Hacettepe University, Faculty of Medicine, Sihhiye, 06100 Ankara, Turkey
| | - Nilufer Sayar
- Department of Molecular Biology and Genetics, Bilkent University, Faculty of Science, Bilkent, 06800 Ankara, Turkey
| | - Turkan Sarioglu
- Department of Histology and Embryology, Istanbul University, Istanbul Medical Faculty, Capa, 34093 Istanbul, Turkey
| | - Ibrahim Oncel
- Department of Pediatrics, Neurology Unit, Hacettepe University, Faculty of Medicine, Sihhiye, 06100 Ankara, Turkey
| | - Parisa Sharafi
- Department of Medical Biology, Hacettepe University, Faculty of Medicine, Sihhiye, 06100 Ankara, Turkey
| | - Hulya Gundesli
- Department of Medical Biology, Hacettepe University, Faculty of Medicine, Sihhiye, 06100 Ankara, Turkey
| | - Burcu Balci-Hayta
- Department of Medical Biology, Hacettepe University, Faculty of Medicine, Sihhiye, 06100 Ankara, Turkey
| | - Nuhan Purali
- Department of Biophysics, Hacettepe University, Faculty of Medicine, Sihhiye, 06100 Ankara, Turkey
| | - Piraye Serdaroglu-Oflazer
- Department of Neurology, Istanbul University, Istanbul Medical Faculty, Capa, 34093 Istanbul, Turkey
| | - Haluk Topaloglu
- Department of Pediatrics, Neurology Unit, Hacettepe University, Faculty of Medicine, Sihhiye, 06100 Ankara, Turkey
| | - Pervin Dincer
- Department of Medical Biology, Hacettepe University, Faculty of Medicine, Sihhiye, 06100 Ankara, Turkey.
| |
Collapse
|