51
|
Khadilkar SV, Yadav RS, Patel BA. Duchenne and Becker Muscular Dystrophies. Neuromuscul Disord 2018. [DOI: 10.1007/978-981-10-5361-0_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
52
|
Soslow JH, Markham LW, Burnette WB, Galindo CL, Feoktistov I, Raucci FJ, Damon BM, Sawyer DB, Ryzhov S. Increased Number of Circulating CD8/CD26 T Cells in the Blood of Duchenne Muscular Dystrophy Patients Is Associated with Augmented Binding of Adenosine Deaminase and Higher Muscular Strength Scores. Front Pharmacol 2017; 8:914. [PMID: 29326589 PMCID: PMC5741593 DOI: 10.3389/fphar.2017.00914] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Accepted: 11/30/2017] [Indexed: 12/02/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked disorder that leads to cardiac and skeletal myopathy. The complex immune activation in boys with DMD is incompletely understood. To better understand the contribution of the immune system into the progression of DMD, we performed a systematic characterization of immune cell subpopulations obtained from peripheral blood of DMD subjects and control donors. We found that the number of CD8 cells expressing CD26 (also known as adenosine deaminase complexing protein 2) was increased in DMD subjects compared to control. No differences, however, were found in the levels of circulating factors associated with pro-inflammatory activation of CD8/CD26 cells, such as tumor necrosis factor-α (TNFα), granzyme B, and interferon-γ (IFNγ). The number of CD8/CD26 cells correlated directly with quantitative muscle testing (QMT) in DMD subjects. Since CD26 mediates binding of adenosine deaminase (ADA) to the T cell surface, we tested ADA-binding capacity of CD8/CD26 cells and the activity of bound ADA. We found that mononuclear cells (MNC) obtained from DMD subjects with an increased number of CD8/CD26 T cells had a greater capacity to bind ADA. In addition, these MNC demonstrated increased hydrolytic deamination of adenosine to inosine. Altogether, our data demonstrated that (1) an increased number of circulating CD8/CD26 T cells is associated with preservation of muscle strength in DMD subjects, and (2) CD8/CD26 T cells from DMD subjects mediated degradation of adenosine by adenosine deaminase. These results support a role for T cells in slowing the decline in skeletal muscle function, and a need for further investigation into contribution of CD8/CD26 T cells in the regulation of chronic inflammation associated with DMD.
Collapse
Affiliation(s)
- Jonathan H Soslow
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Larry W Markham
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States.,Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - W Bryan Burnette
- Division of Pediatric Neurology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Cristi L Galindo
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Igor Feoktistov
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Frank J Raucci
- Thomas P. Graham Jr Division of Pediatric Cardiology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Bruce M Damon
- Departments of Radiology and Radiological Sciences, Molecular Physiology and Biophysics, and Biomedical Engineering, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Douglas B Sawyer
- Maine Medical Center, Portland, ME, United States.,Maine Medical Center Research Institute, Scarborough, ME, United States
| | - Sergey Ryzhov
- Maine Medical Center Research Institute, Scarborough, ME, United States
| |
Collapse
|
53
|
Muscular Dystrophies and Cancer Cachexia: Similarities in Chronic Skeletal Muscle Degeneration. J Funct Morphol Kinesiol 2017. [DOI: 10.3390/jfmk2040039] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
54
|
Pinniger GJ, Terrill JR, Assan EB, Grounds MD, Arthur PG. Pre-clinical evaluation of N-acetylcysteine reveals side effects in the mdx mouse model of Duchenne muscular dystrophy. J Physiol 2017; 595:7093-7107. [PMID: 28887840 DOI: 10.1113/jp274229] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 08/30/2017] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease associated with increased inflammation and oxidative stress. The antioxidant N-acetylcysteine (NAC) has been proposed as a therapeutic intervention for DMD boys, but potential adverse effects of NAC have not been widely investigated. We used young (6 weeks old) growing mdx mice to investigate the capacity of NAC supplementation (2% in drinking water for 6 weeks) to improve dystrophic muscle function and to explore broader systemic effects of NAC treatment. NAC treatment improved normalised measures of muscle function, and decreased inflammation and oxidative stress, but significantly reduced body weight gain, muscle weight and liver weight. Unexpected significant adverse effects of NAC on body and muscle weights indicate that interpretation of muscle function based on normalised force measures should be made with caution and careful consideration is needed when proposing the use of NAC as a therapeutic treatment for young DMD boys. ABSTRACT Duchenne muscular dystrophy (DMD) is a fatal X-linked muscle wasting disease characterised by severe muscle weakness, necrosis, inflammation and oxidative stress. The antioxidant N-acetylcysteine (NAC) has been proposed as a potential therapeutic intervention for DMD boys. We investigated the capacity of NAC to improve dystrophic muscle function in the mdx mouse model of DMD. Young (6 weeks old) mdx and non-dystrophic C57 mice receiving 2% NAC in drinking water for 6 weeks were compared with untreated mice. Grip strength and body weight were measured weekly, before the 12 week old mice were anaesthetised and extensor digitorum longus (EDL) muscles were excised for functional analysis and tissues were sampled for biochemical analyses. Compared to untreated mice, the mean (SD) normalised grip strength was significantly greater in NAC-treated mdx [3.13 (0.58) vs 4.87 (0.78) g body weight (bw)-1 ; P < 0.001] and C57 mice [3.90 (0.32) vs 5.32 (0.60) g bw-1 ; P < 0.001]. Maximum specific force was significantly greater in NAC-treated mdx muscles [9.80 (2.27) vs 13.07 (3.37) N cm-2 ; P = 0.038]. Increased force in mdx mice was associated with reduced thiol oxidation and inflammation in fast muscles, and increased citrate synthase activity in slow muscle. Importantly, NAC significantly impaired body weight gain in both strains of young growing mice, and reduced liver weight in C57 mice and muscle weight in mdx mice. These potentially adverse effects of NAC emphasise the need for caution when interpreting improvements in muscle function based on normalised force measures, and that careful consideration be given to these effects when proposing NAC as a potential treatment for young DMD boys.
Collapse
Affiliation(s)
- Gavin J Pinniger
- School of Human Sciences, the University of Western Australia, Crawley, Western Australia, Australia
| | - Jessica R Terrill
- School of Human Sciences, the University of Western Australia, Crawley, Western Australia, Australia.,School of Molecular Sciences, the University of Western Australia, Perth, Western Australia, Australia
| | - Evanna B Assan
- School of Human Sciences, the University of Western Australia, Crawley, Western Australia, Australia
| | - Miranda D Grounds
- School of Human Sciences, the University of Western Australia, Crawley, Western Australia, Australia
| | - Peter G Arthur
- School of Molecular Sciences, the University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
55
|
Dibenedetto S, Niklison-Chirou M, Cabrera CP, Ellis M, Robson LG, Knopp P, Tedesco FS, Ragazzi M, Di Foggia V, Barnes MR, Radunovic A, Marino S. Enhanced Energetic State and Protection from Oxidative Stress in Human Myoblasts Overexpressing BMI1. Stem Cell Reports 2017; 9:528-542. [PMID: 28735850 PMCID: PMC5549966 DOI: 10.1016/j.stemcr.2017.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 06/17/2017] [Accepted: 06/17/2017] [Indexed: 12/28/2022] Open
Abstract
The Polycomb group gene BMI1 is essential for efficient muscle regeneration in a mouse model of Duchenne muscular dystrophy, and its enhanced expression in adult skeletal muscle satellite cells ameliorates the muscle strength in this model. Here, we show that the impact of mild BMI1 overexpression observed in mouse models is translatable to human cells. In human myoblasts, BMI1 overexpression increases mitochondrial activity, leading to an enhanced energetic state with increased ATP production and concomitant protection against DNA damage both in vitro and upon xenografting in a severe dystrophic mouse model. These preclinical data in mouse models and human cells provide a strong rationale for the development of pharmacological approaches to target BMI1-mediated mitochondrial regulation and protection from DNA damage to sustain the regenerative potential of the skeletal muscle in conditions of chronic muscle wasting.
Collapse
Affiliation(s)
- Silvia Dibenedetto
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Maria Niklison-Chirou
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Claudia P Cabrera
- Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Matthew Ellis
- Division of Neuropathology, the National Hospital for Neurology and Neurosurgery, Queen Square, London WC1N 3BG, UK
| | - Lesley G Robson
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Paul Knopp
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Francesco Saverio Tedesco
- Department of Cell and Developmental Biology, University College London, 21 University Street, London WC1X 0JS, UK
| | - Martina Ragazzi
- Department of Cell and Developmental Biology, University College London, 21 University Street, London WC1X 0JS, UK
| | - Valentina Di Foggia
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK
| | - Michael R Barnes
- Centre for Translational Bioinformatics, William Harvey Research Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Aleksandar Radunovic
- Neuroscience and Trauma Centre, Barts Health NHS Trust, Whitechapel, London E1 1BB, UK
| | - Silvia Marino
- Blizard Institute, Barts and The London School of Medicine and Dentistry, Queen Mary University of London, 4 Newark Street, London E1 2AT, UK.
| |
Collapse
|
56
|
Bhat HF, Mir SS, Dar KB, Bhat ZF, Shah RA, Ganai NA. ABC of multifaceted dystrophin glycoprotein complex (DGC). J Cell Physiol 2017; 233:5142-5159. [DOI: 10.1002/jcp.25982] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 05/01/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Hina F. Bhat
- Division of BiotechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Kashmir SKUAST‐KShuhama, SrinagarJammu and KashmirIndia
| | - Saima S. Mir
- Department of BiotechnologyUniversity of KashmirHazratbal, SrinagarJammu and KashmirIndia
| | - Khalid B. Dar
- Department of BiochemistryUniversity of KashmirHazratbal, SrinagarJammu and KashmirIndia
| | - Zuhaib F. Bhat
- Division of Livestock Products and TechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Jammu (SKUAST‐J), R.S. PoraJammuJammu and KashmirIndia
| | - Riaz A. Shah
- Division of BiotechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Kashmir SKUAST‐KShuhama, SrinagarJammu and KashmirIndia
| | - Nazir A. Ganai
- Division of BiotechnologySher‐e‐Kashmir University of Agricultural Sciences and Technology of Kashmir SKUAST‐KShuhama, SrinagarJammu and KashmirIndia
| |
Collapse
|
57
|
Cellular Reprogramming, Genome Editing, and Alternative CRISPR Cas9 Technologies for Precise Gene Therapy of Duchenne Muscular Dystrophy. Stem Cells Int 2017; 2017:8765154. [PMID: 28607562 PMCID: PMC5451761 DOI: 10.1155/2017/8765154] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/23/2017] [Accepted: 03/28/2017] [Indexed: 01/02/2023] Open
Abstract
In the past decade, the development of two innovative technologies, namely, induced pluripotent stem cells (iPSCs) and the CRISPR Cas9 system, has enabled researchers to model diseases derived from patient cells and precisely edit DNA sequences of interest, respectively. In particular, Duchenne muscular dystrophy (DMD) has been an exemplary monogenic disease model for combining these technologies to demonstrate that genome editing can correct genetic mutations in DMD patient-derived iPSCs. DMD is an X-linked genetic disorder caused by mutations that disrupt the open reading frame of the dystrophin gene, which plays a critical role in stabilizing muscle cells during contraction and relaxation. The CRISPR Cas9 system has been shown to be capable of targeting the dystrophin gene and rescuing its expression in in vitro patient-derived iPSCs and in vivo DMD mouse models. In this review, we highlight recent advances made using the CRISPR Cas9 system to correct genetic mutations and discuss how emerging CRISPR technologies and iPSCs in a combined platform can play a role in bringing a therapy for DMD closer to the clinic.
Collapse
|
58
|
Zhang Q, Liu J, Chen S, Liu J, Liu L, Liu G, Wang F, Jiang W, Zhang C, Wang S, Yuan X. Caspase-12 is involved in stretch-induced apoptosis mediated endoplasmic reticulum stress. Apoptosis 2016; 21:432-42. [PMID: 26801321 DOI: 10.1007/s10495-016-1217-6] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
It is well recognized that mandibular growth, which is caused by a variety of functional appliances, is considered to be the result of both neuromuscular and skeletal adaptations. Accumulating evidence has demonstrated that apoptosis plays an important role in the adaptation of skeletal muscle function. However, the underlying mechanism of apoptosis that is induced by stretch continues to be incompletely understood. Endoplasmic reticulum stress (ERS), a newly defined signaling pathway, initiates apoptosis. This study seeks to determine if caspase-12 is involved in stretch-induced apoptosis mediated endoplasmic reticulum stress in myoblast and its underlying mechanism. Apoptosis was assessed by Hochest staining, DAPI staining and annexin V binding and PI staining. ER chaperones, such as GRP78, CHOP and caspase-12, were determined by reverse transcription polymerase chain reaction (RT-PCR) and Western blot. Furthermore, caspase-12 inhibitor was used to value the mechanism of the caspase-12 pathway. Apoptosis of myoblast, which is subjected to cyclic stretch, was observed in a time-dependent manner. We found that GRP78 mRNA and protein were significantly increased and CHOP and caspase-12 were activated in myoblast that was exposed to cyclic stretch. Caspase-12 inhibition reduced stretch-induced apoptosis, and caspase-12 activated caspase-3 to induce apoptosis. We concluded that caspase-12 played an important role in stretch-induced apoptosis that is associated by endoplasmic reticulum stress by activating caspase-3.
Collapse
Affiliation(s)
- Qiang Zhang
- Department of Orthodontics, Qingdao Municipal Hospital, Qingdao, 266011, Shandong, People's Republic of China
| | - Jianing Liu
- Qingdao Municipal Hospital, Qingdao, 266011, Shandong, People's Republic of China
| | - Shulan Chen
- Qingdao Municipal Hospital, Qingdao, 266011, Shandong, People's Republic of China
| | - Jing Liu
- Qingdao Municipal Hospital, Qingdao, 266011, Shandong, People's Republic of China
| | - Lijuan Liu
- People's of Weifang Hospital, Weifang, 261041, Shandong, People's Republic of China
| | - Guirong Liu
- Department of Orthodontics, Qingdao Municipal Hospital, Qingdao, 266011, Shandong, People's Republic of China
| | - Fang Wang
- Department of Orthodontics, Qingdao Municipal Hospital, Qingdao, 266011, Shandong, People's Republic of China
| | - Wenxin Jiang
- Department of Orthodontics, Qingdao Municipal Hospital, Qingdao, 266011, Shandong, People's Republic of China
| | - Caixia Zhang
- Department of Orthodontics, Qingdao Municipal Hospital, Qingdao, 266011, Shandong, People's Republic of China
| | - Shuangyu Wang
- Department of Stomatology, People's Hospital in Qingdao Shibei District, Qingdao, 266011, Shandong, People's Republic of China
| | - Xiao Yuan
- Department of Orthodontics, Qingdao Municipal Hospital, Qingdao, 266011, Shandong, People's Republic of China.
| |
Collapse
|
59
|
Gonzalez JP, Kyrychenko S, Kyrychenko V, Schneider JS, Granier CJ, Himelman E, Lahey KC, Zhao Q, Yehia G, Tao YX, Bhaumik M, Shirokova N, Fraidenraich D. Small Fractions of Muscular Dystrophy Embryonic Stem Cells Yield Severe Cardiac and Skeletal Muscle Defects in Adult Mouse Chimeras. Stem Cells 2016; 35:597-610. [PMID: 27734557 DOI: 10.1002/stem.2518] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2016] [Revised: 09/10/2016] [Accepted: 09/26/2016] [Indexed: 01/10/2023]
Abstract
Duchenne muscular dystrophy (DMD) is characterized by the loss of the protein dystrophin, leading to muscle fragility, progressive weakening, and susceptibility to mechanical stress. Although dystrophin-negative mdx mouse models have classically been used to study DMD, phenotypes appear mild compared to patients. As a result, characterization of muscle pathology, especially in the heart, has proven difficult. We report that injection of mdx embryonic stem cells (ESCs) into Wild Type blastocysts produces adult mouse chimeras with severe DMD phenotypes in the heart and skeletal muscle. Inflammation, regeneration and fibrosis are observed at the whole organ level, both in dystrophin-negative and dystrophin-positive portions of the chimeric tissues. Skeletal and cardiac muscle function are also decreased to mdx levels. In contrast to mdx heterozygous carriers, which show no significant phenotypes, these effects are even observed in chimeras with low levels of mdx ESC incorporation (10%-30%). Chimeric mice lack typical compensatory utrophin upregulation, and show pathological remodeling of Connexin-43. In addition, dystrophin-negative and dystrophin-positive isolated cardiomyocytes show augmented calcium response to mechanical stress, similar to mdx cells. These global effects highlight a novel role of mdx ESCs in triggering muscular dystrophy even when only low amounts are present. Stem Cells 2017;35:597-610.
Collapse
Affiliation(s)
- J Patrick Gonzalez
- Department of Cell Biology and Molecular Medicine, Newark, New Jersey, USA
| | - Sergii Kyrychenko
- Department of Pharmacology, Physiology and Neuroscience, Newark, New Jersey, USA
| | - Viktoriia Kyrychenko
- Department of Pharmacology, Physiology and Neuroscience, Newark, New Jersey, USA
| | - Joel S Schneider
- Department of Cell Biology and Molecular Medicine, Newark, New Jersey, USA
| | - Celine J Granier
- Department of Pediatrics, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Eric Himelman
- Department of Cell Biology and Molecular Medicine, Newark, New Jersey, USA
| | - Kevin C Lahey
- Department of Cell Biology and Molecular Medicine, Newark, New Jersey, USA
| | - Qingshi Zhao
- Department of Cell Biology and Molecular Medicine, Newark, New Jersey, USA
| | - Ghassan Yehia
- Genome Editing Core Facility, Office of Research Advancement, New Brunswick, New Jersey, USA
| | - Yuan-Xiang Tao
- Department of Cell Biology and Molecular Medicine, Newark, New Jersey, USA.,Department of Pharmacology, Physiology and Neuroscience, Newark, New Jersey, USA.,Department of Anesthesiology, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, New Jersey, USA
| | - Mantu Bhaumik
- Department of Pediatrics, Rutgers Biomedical and Health Sciences, Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Natalia Shirokova
- Department of Pharmacology, Physiology and Neuroscience, Newark, New Jersey, USA
| | - Diego Fraidenraich
- Department of Cell Biology and Molecular Medicine, Newark, New Jersey, USA
| |
Collapse
|
60
|
Spinazzola JM, Kunkel LM. Pharmacological therapeutics targeting the secondary defects and downstream pathology of Duchenne muscular dystrophy. Expert Opin Orphan Drugs 2016; 4:1179-1194. [PMID: 28670506 DOI: 10.1080/21678707.2016.1240613] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Since the identification of the dystrophin gene in 1986, a cure for Duchenne muscular dystrophy (DMD) has yet to be discovered. Presently, there are a number of genetic-based therapies in development aimed at restoration and/or repair of the primary defect. However, growing understanding of the pathophysiological consequences of dystrophin absence has revealed several promising downstream targets for the development of therapeutics. AREAS COVERED In this review, we discuss various strategies for DMD therapy targeting downstream consequences of dystrophin absence including loss of muscle mass, inflammation, fibrosis, calcium overload, oxidative stress, and ischemia. The rationale of each approach and the efficacy of drugs in preclinical and clinical studies are discussed. EXPERT OPINION For the last 30 years, effective DMD drug therapy has been limited to corticosteroids, which are associated with a number of negative side effects. Our knowledge of the consequences of dystrophin absence that contribute to DMD pathology has revealed several potential therapeutic targets. Some of these approaches may have potential to improve or slow disease progression independently or in combination with genetic-based approaches. The applicability of these pharmacological therapies to DMD patients irrespective of their genetic mutation, as well as the potential benefits even for advanced stage patients warrants their continued investigation.
Collapse
Affiliation(s)
- Janelle M Spinazzola
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115
| | - Louis M Kunkel
- Boston Children's Hospital, Division of Genetics and Genomics, Boston, MA 02115.,Harvard Medical School, Departments of Pediatrics and Genetics, Boston, MA 02115.,The Stem Cell Program at Boston Children's Hospital, Boston, MA 02115.,The Manton Center for Orphan Diseases, Boston, MA 02115.,Harvard Stem Cell Institute, Cambridge, MA 02138
| |
Collapse
|
61
|
El-Aloul B, Altamirano-Diaz L, Zapata-Aldana E, Rodrigues R, Malvankar-Mehta MS, Nguyen CT, Campbell C. Pharmacological therapy for the prevention and management of cardiomyopathy in Duchenne muscular dystrophy: A systematic review. Neuromuscul Disord 2016; 27:4-14. [PMID: 27815032 DOI: 10.1016/j.nmd.2016.09.019] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 09/16/2016] [Accepted: 09/26/2016] [Indexed: 01/03/2023]
Abstract
Cardiomyopathy is a major source of morbidity and mortality in Duchenne muscular dystrophy (DMD) patients now that respiratory care has improved. There is currently no definitive evidence guiding the management of DMD-associated cardiomyopathy (DMD-CM). The objective of this systematic review was to evaluate the effectiveness of pharmacotherapies for the prevention and/or management of DMD-CM and to determine the optimal timing to commence these interventions. A systematic search was conducted in January 2016 using MEDLINE, EMBASE and CINAHL databases and grey literature sources for studies evaluating the use of angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers, beta-blockers or aldosterone antagonists. Study quality assessment was conducted using the Downs and Black quality assessment checklist. PRISMA reporting guidelines were used. Of the 15 studies included in this review, most were of low methodological quality. Meta-analysis was not possible due to heterogeneity of studies. ACE inhibitors, angiotensin receptor blockers, beta-blockers and/or aldosterone antagonists tended to improve or preserve left ventricular systolic function and delay the progression of DMD-CM. While there is evidence supporting the use of heart failure medication in patients with DMD, data regarding these interventions for delaying the onset of DMD-CM and when to initiate therapy are lacking. PROSPERO registration: CRD42015029555.
Collapse
Affiliation(s)
- Basmah El-Aloul
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Luis Altamirano-Diaz
- Department of Paediatrics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Eugenio Zapata-Aldana
- Department of Paediatrics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada; Clinical Neurological Sciences, Children's Hospital, London Health Sciences Center, London, ON, Canada
| | - Rebecca Rodrigues
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Monali S Malvankar-Mehta
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada; Department of Ophthalmology, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada
| | - Cam-Tu Nguyen
- Department of Paediatrics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada; Clinical Neurological Sciences, Children's Hospital, London Health Sciences Center, London, ON, Canada
| | - Craig Campbell
- Department of Epidemiology and Biostatistics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada; Department of Paediatrics, Schulich School of Medicine & Dentistry, Western University, London, ON, Canada; Clinical Neurological Sciences, Children's Hospital, London Health Sciences Center, London, ON, Canada.
| |
Collapse
|
62
|
Axonal degeneration, distal collateral branching and neuromuscular junction architecture alterations occur prior to symptom onset in the SOD1G93A mouse model of amyotrophic lateral sclerosis. J Chem Neuroanat 2016; 76:35-47. [DOI: 10.1016/j.jchemneu.2016.03.003] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 01/29/2016] [Accepted: 03/19/2016] [Indexed: 12/11/2022]
|
63
|
Dumont NA, Rudnicki MA. Targeting muscle stem cell intrinsic defects to treat Duchenne muscular dystrophy. NPJ Regen Med 2016; 1. [PMID: 29188075 PMCID: PMC5703417 DOI: 10.1038/npjregenmed.2016.6] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a genetic disease characterised by skeletal muscle degeneration and progressive muscle wasting, which is caused by loss-of-function mutations in the DMD gene that encodes for the protein dystrophin. Dystrophin has critical roles in myofiber stability and integrity by connecting the actin cytoskeleton to the extracellular matrix. Absence of dystrophin leads to myofiber fragility and contributes to skeletal muscle degeneration in DMD patients, however, accumulating evidence also indicate that muscle stem cells (also known as satellite cells) are defective in dystrophic muscles, which leads to impaired muscle regeneration. Our recent work demonstrated that dystrophin is expressed in activated satellite cells, where it regulates the establishment of satellite cell polarity and asymmetric cell division. These findings indicate that dystrophin-deficient satellite cells have intrinsic dysfunctions that contribute to muscle wasting and progression of the disease. This discovery suggests that satellite cells could be targeted to treat DMD. Here we discuss how these new findings affect regenerative therapies for muscular dystrophies. Therapies targeting satellite cells hold great potential and could have long-term efficiency owing to the high self-renewal ability of these cells.
Collapse
Affiliation(s)
- Nicolas A Dumont
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Michael A Rudnicki
- Sprott Center for Stem Cell Research, Ottawa Hospital Research Institute, Regenerative Medicine Program, Ottawa, ON, Canada.,Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
64
|
Duffy RM, Sun Y, Feinberg AW. Understanding the Role of ECM Protein Composition and Geometric Micropatterning for Engineering Human Skeletal Muscle. Ann Biomed Eng 2016; 44:2076-89. [PMID: 26983843 PMCID: PMC4880540 DOI: 10.1007/s10439-016-1592-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/11/2016] [Indexed: 11/27/2022]
Abstract
Skeletal muscle lost through trauma or disease has proven difficult to regenerate due to the challenge of differentiating human myoblasts into aligned, contractile tissue. To address this, we investigated microenvironmental cues that drive myoblast differentiation into aligned myotubes for potential applications in skeletal muscle repair, organ-on-chip disease models and actuators for soft robotics. We used a 2D in vitro system to systematically evaluate the role of extracellular matrix (ECM) protein composition and geometric patterning for controlling the formation of highly aligned myotubes. Specifically, we analyzed myotubes differentiated from murine C2C12 cells and human skeletal muscle derived cells (SkMDCs) on micropatterned lines of laminin compared to fibronectin, collagen type I, and collagen type IV. Results showed that laminin supported significantly greater myotube formation from both cells types, resulting in greater than twofold increase in myotube area on these surfaces compared to the other ECM proteins. Species specific differences revealed that human SkMDCs uniaxially aligned over a wide range of micropatterned line dimensions, while C2C12s required specific line widths and spacings to do the same. Future work will incorporate these results to engineer aligned human skeletal muscle tissue in 2D for in vitro applications in disease modeling, drug discovery and toxicity screening.
Collapse
Affiliation(s)
- Rebecca M Duffy
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, 700 Technology Dr., Pittsburgh, PA, 15219, USA
| | - Yan Sun
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, 700 Technology Dr., Pittsburgh, PA, 15219, USA
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Adam W Feinberg
- Regenerative Biomaterials and Therapeutics Group, Department of Biomedical Engineering, Carnegie Mellon University, 700 Technology Dr., Pittsburgh, PA, 15219, USA.
- Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, 15219, USA.
| |
Collapse
|
65
|
Quaranta MT, Spinello I, Paolillo R, Macchia G, Boe A, Ceccarini M, Labbaye C, Macioce P. Identification of β-Dystrobrevin as a Direct Target of miR-143: Involvement in Early Stages of Neural Differentiation. PLoS One 2016; 11:e0156325. [PMID: 27223470 PMCID: PMC4880309 DOI: 10.1371/journal.pone.0156325] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 05/12/2016] [Indexed: 11/25/2022] Open
Abstract
Duchenne Muscular Dystrophy, a genetic disorder that results in a gradual breakdown of muscle, is associated to mild to severe cognitive impairment in about one-third of dystrophic patients. The brain dysfunction is independent of the muscular pathology, occurs early, and is most likely due to defects in the assembly of the Dystrophin-associated Protein Complex (DPC) during embryogenesis. We have recently described the interaction of the DPC component β-dystrobrevin with members of complexes that regulate chromatin dynamics, and suggested that β-dystrobrevin may play a role in the initiation of neuronal differentiation. Since oxygen concentrations and miRNAs appear as well to be involved in the cellular processes related to neuronal development, we have studied how these factors act on β-dystrobrevin and investigated the possibility of their functional interplay using the NTera-2 cell line, a well-established model for studying neurogenesis. We followed the pattern of expression and regulation of β-dystrobrevin during the early stages of neuronal differentiation induced by exposure to retinoic acid (RA) under hypoxia as compared with normoxia, and found that β-dystrobrevin expression is regulated during RA-induced differentiation of NTera-2 cells. We also found that β-dystrobrevin pattern is delayed under hypoxic conditions, together with a delay in the differentiation and an increase in the proliferation rate of cells. We identified miRNA-143 as a direct regulator of β-dystrobrevin expression, demonstrated that β-dystrobrevin is expressed in the nucleus and showed that, in line with our previous in vitro results, β-dystrobrevin is a repressor of synapsin I in live cells. Altogether the newly identified regulatory pathway miR-143/β-dystrobrevin/synapsin I provides novel insights into the functions of β-dystrobrevin and opens up new perspectives for elucidating the molecular mechanisms underlying the neuronal involvement in muscular dystrophy.
Collapse
Affiliation(s)
- Maria Teresa Quaranta
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Isabella Spinello
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Rosa Paolillo
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Gianfranco Macchia
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Boe
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Marina Ceccarini
- National Centre for Rare Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Catherine Labbaye
- Department of Hematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Rome, Italy
| | - Pompeo Macioce
- Department of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Rome, Italy
- * E-mail:
| |
Collapse
|
66
|
Turk R, Hsiao JJ, Smits MM, Ng BH, Pospisil TC, Jones KS, Campbell KP, Wright ME. Molecular Signatures of Membrane Protein Complexes Underlying Muscular Dystrophy. Mol Cell Proteomics 2016; 15:2169-85. [PMID: 27099343 PMCID: PMC5083101 DOI: 10.1074/mcp.m116.059188] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Indexed: 01/16/2023] Open
Abstract
Mutations in genes encoding components of the sarcolemmal dystrophin-glycoprotein complex (DGC) are responsible for a large number of muscular dystrophies. As such, molecular dissection of the DGC is expected to both reveal pathological mechanisms, and provides a biological framework for validating new DGC components. Establishment of the molecular composition of plasma-membrane protein complexes has been hampered by a lack of suitable biochemical approaches. Here we present an analytical workflow based upon the principles of protein correlation profiling that has enabled us to model the molecular composition of the DGC in mouse skeletal muscle. We also report our analysis of protein complexes in mice harboring mutations in DGC components. Bioinformatic analyses suggested that cell-adhesion pathways were under the transcriptional control of NFκB in DGC mutant mice, which is a finding that is supported by previous studies that showed NFκB-regulated pathways underlie the pathophysiology of DGC-related muscular dystrophies. Moreover, the bioinformatic analyses suggested that inflammatory and compensatory mechanisms were activated in skeletal muscle of DGC mutant mice. Additionally, this proteomic study provides a molecular framework to refine our understanding of the DGC, identification of protein biomarkers of neuromuscular disease, and pharmacological interrogation of the DGC in adult skeletal muscle https://www.mda.org/disease/congenital-muscular-dystrophy/research.
Collapse
Affiliation(s)
- Rolf Turk
- From the ‡Howard Hughes Medical Institute, §Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, ¶Department of Molecular Physiology and Biophysics, ‖Department of Neurology, **Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | | | | | - Brandon H Ng
- ¶Department of Molecular Physiology and Biophysics
| | - Tyler C Pospisil
- From the ‡Howard Hughes Medical Institute, §Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, ¶Department of Molecular Physiology and Biophysics, ‖Department of Neurology, **Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Kayla S Jones
- From the ‡Howard Hughes Medical Institute, §Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, ¶Department of Molecular Physiology and Biophysics, ‖Department of Neurology, **Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | - Kevin P Campbell
- From the ‡Howard Hughes Medical Institute, §Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, ¶Department of Molecular Physiology and Biophysics, ‖Department of Neurology, **Department of Internal Medicine, Roy J. and Lucille A. Carver College of Medicine, The University of Iowa, Iowa City, Iowa
| | | |
Collapse
|
67
|
Hermes TDA, Macedo AB, Fogaça AR, Moraes LHR, de Faria FM, Kido LA, Cagnon VHA, Minatel E. Beneficial cilostazol therapeutic effects inmdxdystrophic skeletal muscle. Clin Exp Pharmacol Physiol 2016; 43:259-67. [DOI: 10.1111/1440-1681.12521] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Revised: 11/17/2015] [Accepted: 11/25/2015] [Indexed: 11/28/2022]
Affiliation(s)
- Túlio de Almeida Hermes
- Department of Structural and Functional Biology; Institute of Biology; State University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Aline Barbosa Macedo
- Department of Structural and Functional Biology; Institute of Biology; State University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Aline Reis Fogaça
- Department of Structural and Functional Biology; Institute of Biology; State University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Luis Henrique Rapucci Moraes
- Department of Structural and Functional Biology; Institute of Biology; State University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Felipe Meira de Faria
- Department of Structural and Functional Biology; Institute of Biology; State University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Larissa Akemi Kido
- Department of Structural and Functional Biology; Institute of Biology; State University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Valéria Helena Alves Cagnon
- Department of Structural and Functional Biology; Institute of Biology; State University of Campinas (UNICAMP); Campinas São Paulo Brazil
| | - Elaine Minatel
- Department of Structural and Functional Biology; Institute of Biology; State University of Campinas (UNICAMP); Campinas São Paulo Brazil
| |
Collapse
|
68
|
Kozakowska M, Pietraszek-Gremplewicz K, Jozkowicz A, Dulak J. The role of oxidative stress in skeletal muscle injury and regeneration: focus on antioxidant enzymes. J Muscle Res Cell Motil 2016; 36:377-93. [PMID: 26728750 PMCID: PMC4762917 DOI: 10.1007/s10974-015-9438-9] [Citation(s) in RCA: 202] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 12/07/2015] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS) are generated in skeletal muscle both during the rest and contractile activity. Myogenic cells are equipped with antioxidant enzymes, like superoxide dismutase, catalase, glutathione peroxidase, γ-glutamylcysteine synthetase and heme oxygenase-1. These enzymes not only neutralise excessive ROS, but also affect myogenic regeneration at several stages: influence post-injury inflammatory reaction, enhance viability and proliferation of muscle satellite cells and myoblasts and affect their differentiation. Finally, antioxidant enzymes regulate also processes accompanying muscle regeneration-induce angiogenesis and reduce fibrosis. Elevated ROS production was also observed in Duchenne muscular dystrophy (DMD), a disease characterised by degeneration of muscle tissue and therefore-increased rate of myogenic regeneration. Antioxidant enzymes are consequently considered as target for therapies counteracting dystrophic symptoms. In this review we present current knowledge regarding the role of oxidative stress and systems of enzymatic antioxidant defence in muscular regeneration after both acute injury and persistent muscular degeneration.
Collapse
Affiliation(s)
- Magdalena Kozakowska
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Katarzyna Pietraszek-Gremplewicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Alicja Jozkowicz
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland.
| | - Jozef Dulak
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Gronostajowa 7, 30-387, Kraków, Poland. .,Malopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland.
| |
Collapse
|
69
|
Rebolledo DL, Kim MJ, Whitehead NP, Adams ME, Froehner SC. Sarcolemmal targeting of nNOSμ improves contractile function of mdx muscle. Hum Mol Genet 2015; 25:158-66. [PMID: 26604149 DOI: 10.1093/hmg/ddv466] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 11/09/2015] [Indexed: 11/13/2022] Open
Abstract
Nitric oxide (NO) is a key regulator of skeletal muscle function and metabolism, including vasoregulation, mitochondrial function, glucose uptake, fatigue and excitation-contraction coupling. The main generator of NO in skeletal muscle is the muscle-specific form of neuronal nitric oxide synthase (nNOSμ) produced by the NOS1 gene. Skeletal muscle nNOSμ is predominantly localized at the sarcolemma by interaction with the dystrophin protein complex (DPC). In Duchenne muscular dystrophy (DMD), loss of dystrophin leads to the mislocalization of nNOSμ from the sarcolemma to the cytosol. This perturbation has been shown to impair contractile function and cause muscle fatigue in dystrophic (mdx) mice. Here, we investigated the effect of restoring sarcolemmal nNOSμ on muscle contractile function in mdx mice. To achieve this, we designed a modified form of nNOSμ (NOS-M) that is targeted to the sarcolemma by palmitoylation, even in the absence of the DPC. When expressed specifically in mdx skeletal muscle, NOS-M significantly attenuates force loss owing to damaging eccentric contractions and repetitive isometric contractions (fatigue), while also improving force recovery after fatigue. Expression of unmodified nNOSμ at similar levels does not lead to sarcolemmal association and fails to improve muscle function. Aside from the benefits of sarcolemmal-localized NO production, NOS-M also increased the surface membrane levels of utrophin and other DPC proteins, including β-dystroglycan, α-syntrophin and α-dystrobrevin in mdx muscle. These results suggest that the expression of NOS-M in skeletal muscle may be therapeutically beneficial in DMD and other muscle diseases characterized by the loss of nNOSμ from the sarcolemma.
Collapse
Affiliation(s)
- Daniela L Rebolledo
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195-7290, USA and Departamento de Biología Celular y Molecular, Pontificia Universidad Católica de Chile, Santiago 8331150, Chile
| | - Min Jeong Kim
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195-7290, USA and
| | - Nicholas P Whitehead
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195-7290, USA and
| | - Marvin E Adams
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195-7290, USA and
| | - Stanley C Froehner
- Department of Physiology and Biophysics, University of Washington, Seattle, WA 98195-7290, USA and
| |
Collapse
|
70
|
Gazzerro E, Baldassari S, Assereto S, Fruscione F, Pistorio A, Panicucci C, Volpi S, Perruzza L, Fiorillo C, Minetti C, Traggiai E, Grassi F, Bruno C. Enhancement of Muscle T Regulatory Cells and Improvement of Muscular Dystrophic Process in mdx Mice by Blockade of Extracellular ATP/P2X Axis. THE AMERICAN JOURNAL OF PATHOLOGY 2015; 185:3349-60. [PMID: 26465071 DOI: 10.1016/j.ajpath.2015.08.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Revised: 08/23/2015] [Accepted: 08/31/2015] [Indexed: 12/19/2022]
Abstract
Infiltration of immune cells and chronic inflammation substantially affect skeletal and cardiac muscle degeneration in Duchenne muscular dystrophy. In the immune system, extracellular adenosine triphosphate (ATP) released by dying cells is sensed as a danger associated molecular pattern through P2 purinergic receptors. Specifically, the P2X7 subtype has a prominent role in regulating immune system physiology and contributes to inflammasome activation also in muscle cells. Here, we show that in vivo blockade of the extracellular ATP/P2X purinergic signaling pathway by periodate-oxidized ATP delayed the progression of the dystrophic phenotype and dampened the local inflammatory response in mdx mice, a spontaneous mouse model of dystrophin deficiency. Reduced infiltration of leukocytes and macrophages and decreased expression of IL-6 were revealed in the muscles of periodate-oxidized ATP-treated mdx mice. Concomitantly, an increase in Foxp3(+) immunosuppressive regulatory T cells was observed and correlated with enhanced myofiber regeneration. Moreover, we detected reduced concentrations of profibrotic cytokines, including transforming growth factor-β and connective tissue growth factor, in muscles of periodate-oxidized ATP-treated mdx mice. The improvement of inflammatory features was associated with increased strength and reduced necrosis, thus suggesting that pharmacologic purinergic antagonism altering the adaptive immune component in the muscle infiltrates might represent a promising therapeutic approach in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Elisabetta Gazzerro
- Unit of Pediatric Neurology and Muscle Disease, Istituto Giannina Gaslini, Genova, Italy
| | - Simona Baldassari
- Unit of Pediatric Neurology and Muscle Disease, Istituto Giannina Gaslini, Genova, Italy
| | - Stefania Assereto
- Unit of Pediatric Neurology and Muscle Disease, Istituto Giannina Gaslini, Genova, Italy
| | - Floriana Fruscione
- Unit of Pediatric Neurology and Muscle Disease, Istituto Giannina Gaslini, Genova, Italy
| | - Angela Pistorio
- Unit of Epidemiology and Statistics, Istituto Giannina Gaslini, Genova, Italy
| | - Chiara Panicucci
- Unit of Pediatric Neurology and Muscle Disease, Istituto Giannina Gaslini, Genova, Italy
| | - Stefano Volpi
- Unit of Pediatrics II, Istituto Giannina Gaslini, Genova, Italy
| | - Lisa Perruzza
- Institute for Research in Biomedicine, Bellinzona, Switzerland; Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
| | - Chiara Fiorillo
- Neuromuscular and Molecular Medicine Unit, Stella Maris Foundation, Pisa, Italy
| | - Carlo Minetti
- Unit of Pediatric Neurology and Muscle Disease, Istituto Giannina Gaslini, Genova, Italy
| | - Elisabetta Traggiai
- Novartis Biologics Center, Novartis Institute for Research in Biomedicine, Basel, Switzerland
| | - Fabio Grassi
- Institute for Research in Biomedicine, Bellinzona, Switzerland; Department of Medical Biotechnologies & Translational Medicine, University of Milan, Istituto Nazionale di Genetica Molecolare, Milan, Italy.
| | - Claudio Bruno
- Center of Myology and Neurodegenerative Disorders, Department of Neuroscience, Istituto Giannina Gaslini, Genova, Italy.
| |
Collapse
|
71
|
Abstract
Duchenne muscular dystrophy, an X-linked disorder, has an incidence of one in 5000 boys and presents in early childhood with proximal muscle weakness. Untreated boys become wheelchair bound by the age of 12 years and die of cardiorespiratory complications in their late teens to early 20s. The use of corticosteroids, non-invasive respiratory support, and active surveillance and management of associated complications have improved ambulation, function, quality of life and life expectancy. The clinical features, investigations and management of Duchenne muscular dystrophy are reviewed, as well as the latest in some of the novel therapies.
Collapse
Affiliation(s)
- Eppie M Yiu
- Neurology Department, Royal Children's Hospital Melbourne, Melbourne, Victoria, Australia.,Bruce Lefroy Centre, Murdoch Childrens Research Institute, Melbourne, Victoria, Australia.,Neurosciences Research, Murdoch Childrens Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| | - Andrew J Kornberg
- Neurology Department, Royal Children's Hospital Melbourne, Melbourne, Victoria, Australia.,Neurosciences Research, Murdoch Childrens Research Institute, Melbourne, Victoria, Australia.,Department of Paediatrics, The University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
72
|
Xu S, Shi D, Pratt SJP, Zhu W, Marshall A, Lovering RM. Abnormalities in brain structure and biochemistry associated with mdx mice measured by in vivo MRI and high resolution localized (1)H MRS. Neuromuscul Disord 2015; 25:764-72. [PMID: 26236031 DOI: 10.1016/j.nmd.2015.07.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 06/21/2015] [Accepted: 07/06/2015] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD), an X-linked disorder caused by the lack of dystrophin, is characterized by the progressive wasting of skeletal muscles. To date, what is known about dystrophin function is derived from studies of dystrophin-deficient animals, with the most common model being the mdx mouse. Most studies on patients with DMD and in mdx mice have focused on skeletal muscle and the development of therapies to reverse, or at least slow, the severe muscle wasting and progressive degeneration. However, dystrophin is also expressed in the CNS. Both mdx mice and patients with DMD can have cognitive and behavioral changes, but studies in the dystrophic brain are limited. We examined the brain structure and metabolites of mature wild type (WT) and mdx mice using magnetic resonance imaging and spectroscopy (MRI/MRS). Both structural and metabolic alterations were observed in the mdx brain. Enlarged lateral ventricles were detected in mdx mice when compared to WT. Diffusion tensor imaging (DTI) revealed elevations in diffusion diffusivities in the prefrontal cortex and a reduction of fractional anisotropy in the hippocampus. Metabolic changes included elevations in phosphocholine and glutathione, and a reduction in γ-aminobutyric acid in the hippocampus. In addition, an elevation in taurine was observed in the prefrontal cortex. Such findings indicate a regional structural change, altered cellular antioxidant defenses, a dysfunction of GABAergic neurotransmission, and a perturbed osmoregulation in the brain lacking dystrophin.
Collapse
Affiliation(s)
- Su Xu
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Da Shi
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Stephen J P Pratt
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Wenjun Zhu
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Andrew Marshall
- Department of Diagnostic Radiology and Nuclear Medicine, School of Medicine, University of Maryland, Baltimore, MD, USA
| | - Richard M Lovering
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, MD, USA.
| |
Collapse
|
73
|
Manning J, O'Malley D. What has the mdx mouse model of Duchenne muscular dystrophy contributed to our understanding of this disease? J Muscle Res Cell Motil 2015; 36:155-67. [PMID: 25669899 DOI: 10.1007/s10974-015-9406-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 01/28/2015] [Indexed: 12/20/2022]
Abstract
Duchenne muscular dystrophy (DMD) is a fatal X-chromosome linked recessive disorder caused by the truncation or deletion of the dystrophin gene. The most widely used animal model of this disease is the dystrophin-deficient mdx mouse which was first discovered 30 years ago. Despite its extensive use in DMD research, no effective treatment has yet been developed for this devastating disease. This review explores what we have learned from this mouse model regarding the pathophysiology of DMD and asks if it has a future in providing a better more thorough understanding of this disease or if it will bring us any closer to improving the outlook for DMD patients.
Collapse
Affiliation(s)
- Jennifer Manning
- Department of Physiology, University College Cork, 4.23 Western Gateway Building, Cork, Ireland
| | | |
Collapse
|
74
|
García-Pelagio KP, Muriel J, O'Neill A, Desmond PF, Lovering RM, Lund L, Bond M, Bloch RJ. Myopathic changes in murine skeletal muscle lacking synemin. Am J Physiol Cell Physiol 2015; 308:C448-62. [PMID: 25567810 DOI: 10.1152/ajpcell.00331.2014] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Diseases of striated muscle linked to intermediate filament (IF) proteins are associated with defects in the organization of the contractile apparatus and its links to costameres, which connect the sarcomeres to the cell membrane. Here we study the role in skeletal muscle of synemin, a type IV IF protein, by examining mice null for synemin (synm-null). Synm-null mice have a mild skeletal muscle phenotype. Tibialis anterior (TA) muscles show a significant decrease in mean fiber diameter, a decrease in twitch and tetanic force, and an increase in susceptibility to injury caused by lengthening contractions. Organization of proteins associated with the contractile apparatus and costameres is not significantly altered in the synm-null. Elastimetry of the sarcolemma and associated contractile apparatus in extensor digitorum longus myofibers reveals a reduction in tension consistent with an increase in sarcolemmal deformability. Although fatigue after repeated isometric contractions is more marked in TA muscles of synm-null mice, the ability of the mice to run uphill on a treadmill is similar to controls. Our results suggest that synemin contributes to linkage between costameres and the contractile apparatus and that the absence of synemin results in decreased fiber size and increased sarcolemmal deformability and susceptibility to injury. Thus synemin plays a moderate but distinct role in fast twitch skeletal muscle.
Collapse
Affiliation(s)
- Karla P García-Pelagio
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Joaquin Muriel
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Andrea O'Neill
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Patrick F Desmond
- Program in Biochemistry and Molecular Biology, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Richard M Lovering
- Department of Orthopaedics, School of Medicine, University of Maryland, Baltimore, Maryland
| | - Linda Lund
- Merrick School of Business, University of Baltimore, Baltimore, Maryland; and
| | - Meredith Bond
- College of Sciences and Health Professions, Cleveland State University, Cleveland, Ohio
| | - Robert J Bloch
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, Maryland;
| |
Collapse
|
75
|
Al-Rewashdy H, Ljubicic V, Lin W, Renaud JM, Jasmin BJ. Utrophin A is essential in mediating the functional adaptations of mdx mouse muscle following chronic AMPK activation. Hum Mol Genet 2014; 24:1243-55. [PMID: 25324540 DOI: 10.1093/hmg/ddu535] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by the absence of dystrophin along muscle fibers. An attractive therapeutic avenue for DMD consists in the upregulation of utrophin A, a protein with high sequence identity and functional redundancy with dystrophin. Recent work has shown that pharmacological interventions that induce a muscle fiber shift toward a slower, more oxidative phenotype with increased expression of utrophin A confer morphological and functional improvements in mdx mice. Whether such improvements result from the increased expression of utrophin A per se or are linked to other beneficial adaptations associated with the slow, oxidative phenotype remain to be established. To address this central issue, we capitalized on the use of double knockout (dKO) mice, which are mdx mice also deficient in utrophin. We first compared expression of signaling molecules and markers of the slow, oxidative phenotype in muscles of mdx versus dKO mice and found that both strains exhibit similar phenotypes. Chronic activation of 5' adenosine monophosphate-activated protein kinase with 5-amino-4-imidazolecarboxamide riboside (AICAR) resulted in expression of a slower, more oxidative phenotype in both mdx and dKO mice. In mdx mice, this fiber type shift was accompanied by clear functional improvements that included reductions in central nucleation, IgM sarcoplasmic penetration and sarcolemmal damage resulting from eccentric contractions, as well as in increased grip strength. These important morphological and functional adaptations were not seen in AICAR-treated dKO mice. Our findings show the central role of utrophin A in mediating the functional benefits associated with expression of a slower, more oxidative phenotype in dystrophic animals.
Collapse
Affiliation(s)
- Hasanen Al-Rewashdy
- Department of Cellular and Molecular Medicine, and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Vladimir Ljubicic
- Department of Cellular and Molecular Medicine, and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Wei Lin
- Department of Cellular and Molecular Medicine, and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Jean-Marc Renaud
- Department of Cellular and Molecular Medicine, and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| | - Bernard J Jasmin
- Department of Cellular and Molecular Medicine, and Centre for Neuromuscular Disease, Faculty of Medicine, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
76
|
Ogura Y, Tajrishi MM, Sato S, Hindi SM, Kumar A. Therapeutic potential of matrix metalloproteinases in Duchenne muscular dystrophy. Front Cell Dev Biol 2014; 2:11. [PMID: 25364719 PMCID: PMC4207008 DOI: 10.3389/fcell.2014.00011] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 03/13/2014] [Indexed: 12/31/2022] Open
Abstract
Matrix metalloproteinases (MMPs) are secreted proteinases that have physiologic roles in degradation and remodeling of extracellular matrix (ECM) in almost all tissues. However, their excessive production in disease conditions leads to many pathological features including tissue breakdown, inflammation, cell death, and fibrosis. Duchenne Muscular dystrophy (DMD) is a devastating genetic muscle disorder caused by partial or complete loss of cytoskeletal protein dystrophin. Progressive muscle wasting in DMD is accompanied by myofiber necrosis followed by cycles of regeneration and degeneration and inflammation that eventually result in replacement of myofiber by connective and adipose tissues. Emerging evidence suggests that gene expression and the activity of various MMPs are aberrantly regulated in muscle biopsies from DMD patients and in skeletal muscle of animal models of DMD. Moreover, a few studies employing genetic mouse models have revealed that different MMPs play distinct roles in disease progression in DMD. Modulation of the activity of MMPs improves myofiber regeneration and enhances the efficacy of transplantation and engraftment of muscle progenitor cells in dystrophic muscle in mouse models of DMD. Furthermore, recent reports also suggest that some MMPs especially MMP-9 can serve as a biomarker for diagnosis and prognosis of DMD. In this article, we provide a succinct overview of the regulation of various MMPs and their therapeutic importance in DMD.
Collapse
Affiliation(s)
- Yuji Ogura
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| | - Marjan M Tajrishi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| | - Shuichi Sato
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| | - Sajedah M Hindi
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| | - Ashok Kumar
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| |
Collapse
|
77
|
Valladares D, Almarza G, Contreras A, Pavez M, Buvinic S, Jaimovich E, Casas M. Electrical stimuli are anti-apoptotic in skeletal muscle via extracellular ATP. Alteration of this signal in Mdx mice is a likely cause of dystrophy. PLoS One 2013; 8:e75340. [PMID: 24282497 PMCID: PMC3839923 DOI: 10.1371/journal.pone.0075340] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Accepted: 08/11/2013] [Indexed: 12/20/2022] Open
Abstract
ATP signaling has been shown to regulate gene expression in skeletal muscle and to be altered in models of muscular dystrophy. We have previously shown that in normal muscle fibers, ATP released through Pannexin1 (Panx1) channels after electrical stimulation plays a role in activating some signaling pathways related to gene expression. We searched for a possible role of ATP signaling in the dystrophy phenotype. We used muscle fibers from flexor digitorum brevis isolated from normal and mdx mice. We demonstrated that low frequency electrical stimulation has an anti-apoptotic effect in normal muscle fibers repressing the expression of Bax, Bim and PUMA. Addition of exogenous ATP to the medium has a similar effect. In dystrophic fibers, the basal levels of extracellular ATP were higher compared to normal fibers, but unlike control fibers, they do not present any ATP release after low frequency electrical stimulation, suggesting an uncoupling between electrical stimulation and ATP release in this condition. Elevated levels of Panx1 and decreased levels of Cav1.1 (dihydropyridine receptors) were found in triads fractions prepared from mdx muscles. Moreover, decreased immunoprecipitation of Cav1.1 and Panx1, suggest uncoupling of the signaling machinery. Importantly, in dystrophic fibers, exogenous ATP was pro-apoptotic, inducing the transcription of Bax, Bim and PUMA and increasing the levels of activated Bax and cytosolic cytochrome c. These evidence points to an involvement of the ATP pathway in the activation of mechanisms related with cell death in muscular dystrophy, opening new perspectives towards possible targets for pharmacological therapies.
Collapse
Affiliation(s)
- Denisse Valladares
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Gonzalo Almarza
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Ariel Contreras
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mario Pavez
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sonja Buvinic
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Departamento de Ciencias Básicas y Comunitarias, Facultad de Odontología, Universidad de Chile, Santiago, Chile
| | - Enrique Jaimovich
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Mariana Casas
- Centro de Estudios Moleculares de la Célula, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Santiago, Chile
- * E-mail:
| |
Collapse
|
78
|
Gao HL, Li C, Nabeka H, Shimokawa T, Kobayashi N, Saito S, Wang ZY, Cao YM, Matsuda S. Decrease in prosaposin in the Dystrophic mdx mouse brain. PLoS One 2013; 8:e80032. [PMID: 24244600 PMCID: PMC3828254 DOI: 10.1371/journal.pone.0080032] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Accepted: 09/27/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy caused by a mutation in the X-linked dystrophin gene induces metabolic and structural disorders in the brain. A lack of dystrophin in brain structures is involved in impaired cognitive function. Prosaposin (PS), a neurotrophic factor, is abundant in the choroid plexus and various brain regions. We investigated whether PS serves as a link between dystrophin loss and gross and/or ultrastructural brain abnormalities. METHODOLOGY/PRINCIPAL FINDINGS The distribution of PS in the brains of juvenile and adult mdx mice was investigated by immunochemistry, Western blotting, and in situ hybridization. Immunochemistry revealed lower levels of PS in the cytoplasm of neurons of the cerebral cortex, hippocampus, cerebellum, and choroid plexus in mdx mice. Western blotting confirmed that PS levels were lower in these brain regions in both juveniles and adults. Even with low PS production in the choroids plexus, there was no significant PS decrease in cerebrospinal fluid (CSF). In situ hybridization revealed that the primary form of PS mRNA in both normal and mdx mice was Pro+9, a secretory-type PS, and the hybridization signals for Pro+9 in the above-mentioned brain regions were weaker in mdx mice than in normal mice. We also investigated mitogen-activated protein kinase signalling. Stronger activation of ERK1/2 was observed in mdx mice, ERK1/2 activity was positively correlated with PS activity, and exogenous PS18 stimulated both p-ERK1/2 and PS in SH-SY5Y cells. CONCLUSIONS/SIGNIFICANCE Low levels of PS and its receptors suggest the participation of PS in some pathological changes in the brains of mdx mice.
Collapse
Affiliation(s)
- Hui-ling Gao
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Cheng Li
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
- Department of Immunology, China Medical University, Shenyang, China
| | - Hiroaki Nabeka
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Tetsuya Shimokawa
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Naoto Kobayashi
- Medical Education Center, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| | - Shouichiro Saito
- Laboratory of Veterinary Anatomy, Faculty of Applied Biological Sciences, Gifu University, Yanagido, Gifu, Japan
| | - Zhan-You Wang
- College of Life and Health Sciences, Northeastern University, Shenyang, China
| | - Ya-ming Cao
- Department of Immunology, China Medical University, Shenyang, China
| | - Seiji Matsuda
- Department of Anatomy and Embryology, Ehime University Graduate School of Medicine, Toon, Ehime, Japan
| |
Collapse
|
79
|
Transcriptional networks regulating the costamere, sarcomere, and other cytoskeletal structures in striated muscle. Cell Mol Life Sci 2013; 71:1641-56. [PMID: 24218011 DOI: 10.1007/s00018-013-1512-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2013] [Revised: 10/27/2013] [Accepted: 10/30/2013] [Indexed: 10/26/2022]
Abstract
Structural abnormalities in striated muscle have been observed in numerous transcription factor gain- and loss-of-function phenotypes in animal and cell culture model systems, indicating that transcription is important in regulating the cytoarchitecture. While most characterized cytoarchitectural defects are largely indistinguishable by histological and ultrastructural criteria, analysis of dysregulated gene expression in each mutant phenotype has yielded valuable information regarding specific structural gene programs that may be uniquely controlled by each of these transcription factors. Linking the formation and maintenance of each subcellular structure or subset of proteins within a cytoskeletal compartment to an overlapping but distinct transcription factor cohort may enable striated muscle to control cytoarchitectural function in an efficient and specific manner. Here we summarize the available evidence that connects transcription factors, those with established roles in striated muscle such as MEF2 and SRF, as well as other non-muscle transcription factors, to the regulation of a defined cytoskeletal structure. The notion that genes encoding proteins localized to the same subcellular compartment are coordinately transcriptionally regulated may prompt rationally designed approaches that target specific transcription factor pathways to correct structural defects in muscle disease.
Collapse
|
80
|
Hindi SM, Sato S, Choi Y, Kumar A. Distinct roles of TRAF6 at early and late stages of muscle pathology in the mdx model of Duchenne muscular dystrophy. Hum Mol Genet 2013; 23:1492-505. [PMID: 24163132 DOI: 10.1093/hmg/ddt536] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal genetic disorder caused by loss of functional dystrophin protein. Accumulating evidence suggests that the deficiency of dystrophin leads to aberrant activation of many signaling pathways which contribute to disease progression. However, the proximal signaling events leading to the activation of various pathological cascades in dystrophic muscle remain less clear. TNF receptor-associated factor 6 (TRAF6) is an adaptor protein which acts as a signaling intermediate for several receptor-mediated signaling events leading to the context-dependent activation of a number of signaling pathways. TRAF6 is also an E3 ubiquitin ligase and an important regulator of autophagy. However, the role of TRAF6 in pathogenesis of DMD remains unknown. Here, we demonstrate that the levels and activity of TRAF6 are increased in skeletal muscle of mdx (a mouse model of DMD) mice. Targeted deletion of TRAF6 improves muscle strength and reduces fiber necrosis, infiltration of macrophages and the activation of proinflammatory transcription factor nuclear factor-kappa B (NF-κB) in 7-week-old mdx mice. Ablation of TRAF6 also increases satellite cells proliferation and myofiber regeneration in young mdx mice. Intriguingly, ablation of TRAF6 exacerbates muscle injury and increases fibrosis in 9-month-old mdx mice. TRAF6 inhibition reduces the markers of autophagy and Akt signaling in dystrophic muscle of mdx mice. Collectively, our study suggests that while the inhibition of TRAF6 improves muscle structure and function in young mdx mice, its continued inhibition causes more severe myopathy at later stages of disease progression potentially through repressing autophagy.
Collapse
Affiliation(s)
- Sajedah M Hindi
- Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA and
| | | | | | | |
Collapse
|
81
|
Marti M, Montserrat N, Pardo C, Mulero L, Miquel-Serra L, Rodrigues AMC, Andrés Vaquero J, Kuebler B, Morera C, Barrero MJ, Izpisua Belmonte JC. M-cadherin-mediated intercellular interactions activate satellite cell division. J Cell Sci 2013; 126:5116-31. [PMID: 24046443 DOI: 10.1242/jcs.123562] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Adult muscle stem cells and their committed myogenic precursors, commonly referred to as the satellite cell population, are involved in both muscle growth after birth and regeneration after damage. It has been previously proposed that, under these circumstances, satellite cells first become activated, divide and differentiate, and only later fuse to the existing myofiber through M-cadherin-mediated intercellular interactions. Our data show that satellite cells fuse with the myofiber concomitantly to cell division, and only when the nuclei of the daughter cells are inside the myofiber, do they complete the process of differentiation. Here we demonstrate that M-cadherin plays an important role in cell-to-cell recognition and fusion, and is crucial for cell division activation. Treatment of satellite cells with M-cadherin in vitro stimulates cell division, whereas addition of anti-M-cadherin antibodies reduces the cell division rate. Our results suggest an alternative model for the contribution of satellite cells to muscle development, which might be useful in understanding muscle regeneration, as well as muscle-related dystrophies.
Collapse
Affiliation(s)
- Merce Marti
- Center of Regenerative Medicine in Barcelona, Dr. Aiguader, 88, 08003 Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Dick E, Kalra S, Anderson D, George V, Ritso M, Laval SH, Barresi R, Aartsma-Rus A, Lochmüller H, Denning C. Exon skipping and gene transfer restore dystrophin expression in human induced pluripotent stem cells-cardiomyocytes harboring DMD mutations. Stem Cells Dev 2013; 22:2714-24. [PMID: 23829870 DOI: 10.1089/scd.2013.0135] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
With an incidence of ∼1:3,500 to 5,000 in male children, Duchenne muscular dystrophy (DMD) is an X-linked disorder in which progressive muscle degeneration occurs and affected boys usually die in their twenties or thirties. Cardiac involvement occurs in 90% of patients and heart failure accounts for up to 40% of deaths. To enable new therapeutics such as gene therapy and exon skipping to be tested in human cardiomyocytes, we produced human induced pluripotent stem cells (hiPSC) from seven patients harboring mutations across the DMD gene. Mutations were retained during differentiation and analysis indicated the cardiomyocytes showed a dystrophic gene expression profile. Antisense oligonucleotide-mediated skipping of exon 51 restored dystrophin expression to ∼30% of normal levels in hiPSC-cardiomyocytes carrying exon 47-50 or 48-50 deletions. Alternatively, delivery of a dystrophin minigene to cardiomyocytes with a deletion in exon 35 or a point mutation in exon 70 allowed expression levels similar to those seen in healthy cells. This demonstrates that DMD hiPSC-cardiomyocytes provide a novel tool to evaluate whether new therapeutics can restore dystrophin expression in the heart.
Collapse
Affiliation(s)
- Emily Dick
- 1 Wolfson Centre for Stem Cells, Tissue Engineering and Modelling (STEM), Centre for Biomolecular Sciences, University of Nottingham , Nottingham, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Giacomotto J, Brouilly N, Walter L, Mariol MC, Berger J, Ségalat L, Becker TS, Currie PD, Gieseler K. Chemical genetics unveils a key role of mitochondrial dynamics, cytochrome c release and IP3R activity in muscular dystrophy. Hum Mol Genet 2013; 22:4562-78. [PMID: 23804750 DOI: 10.1093/hmg/ddt302] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is a neuromuscular disease caused by mutations in the dystrophin gene. The subcellular mechanisms of DMD remain poorly understood and there is currently no curative treatment available. Using a Caenorhabditis elegans model for DMD as a pharmacologic and genetic tool, we found that cyclosporine A (CsA) reduces muscle degeneration at low dose and acts, at least in part, through a mitochondrial cyclophilin D, CYN-1. We thus hypothesized that CsA acts on mitochondrial permeability modulation through cyclophilin D inhibition. Mitochondrial patterns and dynamics were analyzed, which revealed dramatic mitochondrial fragmentation not only in dystrophic nematodes, but also in a zebrafish model for DMD. This abnormal mitochondrial fragmentation occurs before any obvious sign of degeneration can be detected. Moreover, we demonstrate that blocking/delaying mitochondrial fragmentation by knocking down the fission-promoting gene drp-1 reduces muscle degeneration and improves locomotion abilities of dystrophic nematodes. Further experiments revealed that cytochrome c is involved in muscle degeneration in C. elegans and seems to act, at least in part, through an interaction with the inositol trisphosphate receptor calcium channel, ITR-1. Altogether, our findings reveal that mitochondria play a key role in the early process of muscle degeneration and may be a target of choice for the design of novel therapeutics for DMD. In addition, our results provide the first indication in the nematode that (i) mitochondrial permeability transition can occur and (ii) cytochrome c can act in cell death.
Collapse
Affiliation(s)
- Jean Giacomotto
- Brain and Mind Research Institute, Sydney Medical School, University of Sydney, NSW 2050, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
84
|
Frank CA. Homeostatic plasticity at the Drosophila neuromuscular junction. Neuropharmacology 2013; 78:63-74. [PMID: 23806804 DOI: 10.1016/j.neuropharm.2013.06.015] [Citation(s) in RCA: 114] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Revised: 05/31/2013] [Accepted: 06/11/2013] [Indexed: 02/07/2023]
Abstract
In biology, homeostasis refers to how cells maintain appropriate levels of activity. This concept underlies a balancing act in the nervous system. Synapses require flexibility (i.e. plasticity) to adjust to environmental challenges. Yet there must also exist regulatory mechanisms that constrain activity within appropriate physiological ranges. An abundance of evidence suggests that homeostatic regulation is critical in this regard. In recent years, important progress has been made toward identifying molecules and signaling processes required for homeostatic forms of neuroplasticity. The Drosophila melanogaster third instar larval neuromuscular junction (NMJ) has been an important experimental system in this effort. Drosophila neuroscientists combine genetics, pharmacology, electrophysiology, imaging, and a variety of molecular techniques to understand how homeostatic signaling mechanisms take shape at the synapse. At the NMJ, homeostatic signaling mechanisms couple retrograde (muscle-to-nerve) signaling with changes in presynaptic calcium influx, changes in the dynamics of the readily releasable vesicle pool, and ultimately, changes in presynaptic neurotransmitter release. Roles in these processes have been demonstrated for several molecules and signaling systems discussed here. This review focuses primarily on electrophysiological studies or data. In particular, attention is devoted to understanding what happens when NMJ function is challenged (usually through glutamate receptor inhibition) and the resulting homeostatic responses. A significant area of study not covered in this review, for the sake of simplicity, is the homeostatic control of synapse growth, which naturally, could also impinge upon synapse function in myriad ways. This article is part of the Special Issue entitled 'Homeostatic Synaptic Plasticity'.
Collapse
Affiliation(s)
- C Andrew Frank
- Department of Anatomy and Cell Biology, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA.
| |
Collapse
|
85
|
Shin J, Tajrishi MM, Ogura Y, Kumar A. Wasting mechanisms in muscular dystrophy. Int J Biochem Cell Biol 2013; 45:2266-79. [PMID: 23669245 DOI: 10.1016/j.biocel.2013.05.001] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 04/29/2013] [Accepted: 05/02/2013] [Indexed: 12/11/2022]
Abstract
Muscular dystrophy is a group of more than 30 different clinical genetic disorders that are characterized by progressive skeletal muscle wasting and degeneration. Primary deficiency of specific extracellular matrix, sarcoplasmic, cytoskeletal, or nuclear membrane protein results in several secondary changes such as sarcolemmal instability, calcium influx, fiber necrosis, oxidative stress, inflammatory response, breakdown of extracellular matrix, and eventually fibrosis which leads to loss of ambulance and cardiac and respiratory failure. A number of molecular processes have now been identified which hasten disease progression in human patients and animal models of muscular dystrophy. Accumulating evidence further suggests that aberrant activation of several signaling pathways aggravate pathological cascades in dystrophic muscle. Although replacement of defective gene with wild-type is paramount to cure, management of secondary pathological changes has enormous potential to improving the quality of life and extending lifespan of muscular dystrophy patients. In this article, we have reviewed major cellular and molecular mechanisms leading to muscle wasting in muscular dystrophy. This article is part of a Directed Issue entitled: Molecular basis of muscle wasting.
Collapse
Affiliation(s)
- Jonghyun Shin
- Department of Anatomical Sciences and Neurobiology, University of Louisville School of Medicine, Louisville, KY 40202, USA
| | | | | | | |
Collapse
|
86
|
Abstract
Diseases of muscle may be congenital or acquired. They cause muscle weakness without sensory loss. The onset, distribution, and clinical course help to differentiate the type of muscle disorder. The diagnostic workup may include laboratory examination, electrodiagnostic studies, and muscle biopsy. A definitive diagnosis leads to better decision making with regard to treatment, genetic education, prognosis, functional expectations, and the impact of exercise on muscle function.
Collapse
Affiliation(s)
- Anthony Chiodo
- Physical Medicine and Rehabilitation, University of Michigan Hospital, 325 E Eisenhower Parkway, Ann Arbor, MI 48118, USA.
| |
Collapse
|
87
|
Soltow QA, Zeanah EH, Lira VA, Criswell DS. Cessation of cyclic stretch induces atrophy of C2C12 myotubes. Biochem Biophys Res Commun 2013; 434:316-21. [PMID: 23541574 DOI: 10.1016/j.bbrc.2013.03.048] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2013] [Accepted: 03/15/2013] [Indexed: 10/27/2022]
Abstract
Cyclic stretch of differentiated myotubes mimics the loading pattern of mature skeletal muscle. We tested a cell culture model of disuse atrophy by the cessation of repetitive bouts of cyclic stretch in differentiated C2C12 myotubes. Myotubes were subjected to cyclic strain (12%, 0.7 Hz, 1 h/d) on collagen-I-coated Bioflex plates using a computer-controlled vacuum stretch apparatus (Flexcell Int.) for 2 (2dSTR) or 5 (5dSTR) consecutive days. Control cultures were maintained in the Bioflex plates without cyclic stretch for 2d or 5d. Additionally, some cultures were stretched for 2 d followed by cessation of stretch for 3d (2dSTR3dCES). Cyclic stretching (5dSTR) increased myotube diameter and overall myotube area by ~2-fold (P<0.05) compared to non-stretched controls, while cessation of stretch (2dSTR3dCES) resulted in ~80% smaller myotubes than 5dSTR cells, and 40-50% smaller than non-stretched controls (P<0.05). Further, the calpain-dependent cleavage products of αII-spectrin (150 kDa) and talin increased (3.5-fold and 2.2-fold, respectively; P<0.05) in 2dSTR3dCES myotubes, compared to non-stretched controls. The 1h cyclic stretching protocol acutely increased the phosphorylation of Akt (+4.5-fold; P<0.05) and its downstream targets, FOXO3a (+4.2-fold; P<0.05) and GSK-3β (+1.8-fold; P<0.05), which returned to baseline by 48 h after cessation of stretch. Additionally, nitric oxide production increased during stretch and co-treatment with the NOS inhibitor, l-NAME, inhibited the effects of stretch and cessation of stretch. We conclude that cessation of cyclic stretching causes myotube atrophy by activating calpains and decreasing activation of Akt. Stretch-induced myotube growth, as well as activation of atrophy signaling with cessation of stretch, are dependent on NOS activity.
Collapse
Affiliation(s)
- Quinlyn A Soltow
- Center for Exercise Science, Department of Applied Physiology & Kinesiology, University of Florida, Gainesville, FL 32611, USA
| | | | | | | |
Collapse
|
88
|
Nedergaard A, Karsdal MA, Sun S, Henriksen K. Serological muscle loss biomarkers: an overview of current concepts and future possibilities. J Cachexia Sarcopenia Muscle 2013; 4:1-17. [PMID: 22996343 PMCID: PMC3581612 DOI: 10.1007/s13539-012-0086-2] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2012] [Accepted: 08/21/2012] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The skeletal muscle mass is the largest organ in the healthy body, comprising 30-40 % of the body weight of an adult man. It confers protection from trauma, locomotion, ventilation, and it represents a "sink" in glucose metabolism and a reservoir of amino acids to other tissues such as the brain and blood cells. Naturally, loss of muscle has dire consequences for health as well as functionality. Muscle loss is a natural consequence of especially aging, inactivity, and their associated metabolic dysfunction, but it is strongly accelerated in critical illness such as organ failure, sepsis, or cancer. Whether this muscle loss is considered a primary or secondary condition, it is known that muscle loss is a symptom that predicts morbidity and mortality and one that is known to impact quality of life and independence. Therefore, monitoring of muscle mass is relevant in a number of pathologies as well as in clinical trials as measures of efficacy as well as safety. METHODS AND RESULTS Existing biomarkers of muscle mass or muscle loss have shown to be either too unreliable or too impractical in relation to the perceived clinical benefit to reach regular clinical research or use. We suggest serological neoepitope biomarkers as a possible technology to address some of these problems. Blood biomarkers of this kind have previously been shown to respond with high sensitivity and shorter time to minimum significant change than available biomarkers of muscle mass. We provide brief reviews of existing muscle mass or function biomarker technologies, muscle protein biology, and existing neoepitope biomarkers and proceed to present tentative recommendations on how to select and detect neoepitope biomarkers. CONCLUSION We suggest that serological peptide biomarkers whose tissue and pathology specificity are derived from post-translational modification of proteins in tissues of interest, presenting so-called neoepitopes, represents an exciting candidate technology to fill out an empty niche in biomarker technology.
Collapse
|
89
|
Martínez-Vieyra IA, Vásquez-Limeta A, González-Ramírez R, Morales-Lázaro SL, Mondragón M, Mondragón R, Ortega A, Winder SJ, Cisneros B. A role for β-dystroglycan in the organization and structure of the nucleus in myoblasts. BIOCHIMICA ET BIOPHYSICA ACTA 2013; 1833:698-711. [PMID: 23220011 DOI: 10.1016/j.bbamcr.2012.11.019] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2012] [Revised: 11/17/2012] [Accepted: 11/24/2012] [Indexed: 12/19/2022]
Abstract
We recently characterized a nuclear import pathway for β-dystroglycan; however, its nuclear role remains unknown. In this study, we demonstrate for the first time, the interaction of β-dystroglycan with distinct proteins from different nuclear compartments, including the nuclear envelope (NE) (emerin and lamins A/C and B1), splicing speckles (SC35), Cajal bodies (p80-coilin), and nucleoli (Nopp140). Electron microscopy analysis revealed that β-dystroglycan localized in the inner nuclear membrane, nucleoplasm, and nucleoli. Interestingly, downregulation of β-dystroglycan resulted in both mislocalization and decreased expression of emerin and lamin B1, but not lamin A/C, as well in disorganization of nucleoli, Cajal bodies, and splicing speckles with the concomitant decrease in the levels of Nopp140, and p80-coilin, but not SC35. Quantitative reverse transcription PCR and cycloheximide-mediated protein arrest assays revealed that β-dystroglycan deficiency did not change mRNA expression of NE proteins emerin and lamin B1 bud did alter their stability, accelerating protein turnover. Furthermore, knockdown of β-dystroglycan disrupted NE-mediated processes including nuclear morphology and centrosome-nucleus linkage, which provides evidence that β-dystroglycan association with NE proteins is biologically relevant. Unexpectedly, β-dystroglycan-depleted cells exhibited multiple centrosomes, a characteristic of cancerous cells. Overall, these findings imply that β-dystroglycan is a nuclear scaffolding protein involved in nuclear organization and NE structure and function, and that might be a contributor to the biogenesis of nuclear envelopathies.
Collapse
Affiliation(s)
- Ivette A Martínez-Vieyra
- Departamento de Genética y Biología Molecular, Centro de Investigación y de Estudios Avanzados del IPN, México, DF 07360, Mexico
| | | | | | | | | | | | | | | | | |
Collapse
|
90
|
Pertl C, Eblenkamp M, Pertl A, Pfeifer S, Wintermantel E, Lochmüller H, Walter MC, Krause S, Thirion C. A new web-based method for automated analysis of muscle histology. BMC Musculoskelet Disord 2013; 14:26. [PMID: 23324401 PMCID: PMC3560198 DOI: 10.1186/1471-2474-14-26] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2012] [Accepted: 01/13/2013] [Indexed: 11/10/2022] Open
Abstract
Background Duchenne Muscular Dystrophy is an inherited degenerative neuromuscular disease characterised by rapidly progressive muscle weakness. Currently, curative treatment is not available. Approaches for new treatments that improve muscle strength and quality of life depend on preclinical testing in animal models. The mdx mouse model is the most frequently used animal model for preclinical studies in muscular dystrophy research. Standardised pathology-relevant parameters of dystrophic muscle in mdx mice for histological analysis have been developed in international, collaborative efforts, but automation has not been accessible to most research groups. A standardised and mainly automated quantitative assessment of histopathological parameters in the mdx mouse model is desirable to allow an objective comparison between laboratories. Methods Immunological and histochemical reactions were used to obtain a double staining for fast and slow myosin. Additionally, fluorescence staining of the myofibre membranes allows defining the minimal Feret’s diameter. The staining of myonuclei with the fluorescence dye bisbenzimide H was utilised to identify nuclei located internally within myofibres. Relevant structures were extracted from the image as single objects and assigned to different object classes using web-based image analysis (MyoScan). Quantitative and morphometric data were analysed, e.g. the number of nuclei per fibre and minimal Feret’s diameter in 6 month old wild-type C57BL/10 mice and mdx mice. Results In the current version of the module “MyoScan”, essential parameters for histologic analysis of muscle sections were implemented including the minimal Feret’s diameter of the myofibres and the automated calculation of the percentage of internally nucleated myofibres. Morphometric data obtained in the present study were in good agreement with previously reported data in the literature and with data obtained from manual analysis. Conclusions A standardised and mainly automated quantitative assessment of histopathological parameters in the mdx mouse model is now available. Automated analysis of histological parameters is more rapid and less time-consuming. Moreover, results are unbiased and more reliable. Efficacy of therapeutic interventions, e.g. within the scope of a drug screening or therapeutic exon skipping, can be monitored. The automatic analysis system MyoScan used in this study is not limited exclusively to dystrophin-deficient mice but also represents a useful tool for applications in the research of other dystrophic pathologies, various other skeletal muscle diseases and degenerative neuromuscular disorders.
Collapse
Affiliation(s)
- Cordula Pertl
- Laboratory of Molecular Myology, Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-Universität München, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Sharma A, Sane H, Badhe P, Gokulchandran N, Kulkarni P, Lohiya M, Biju H, Jacob VC. A Clinical Study Shows Safety and Efficacy of Autologous Bone Marrow Mononuclear Cell Therapy to Improve Quality of Life in Muscular Dystrophy Patients. Cell Transplant 2013; 22 Suppl 1:S127-38. [DOI: 10.3727/096368913x672136] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Muscular dystrophy is a genetic disorder with no definite cure. A study was carried out on 150 patients diagnosed with muscular dystrophy. These included Duchenne muscular dystrophy, limb-girdle muscular dystrophy, and Becker muscular dystrophy variants. They were administered autologous bone marrow-derived mononuclear cells intrathecally and intramuscularly at the motor points of the antigravity weak muscles followed by vigorous rehabilitation therapy. No significant adverse events were noted. Assessment after transplantation showed neurological improvements in trunk muscle strength, limb strength on manual muscle testing, gait improvements, and a favorable shift on assessment scales such as the Functional Independence Measure and the Brooke and Vignos Scales. Furthermore, imaging and electrophysiological studies also showed significant changes in selective cases. On a mean follow-up of 12 ± 1 months, overall 86.67% cases showed symptomatic and functional improvements, with six patients showing changes with respect to muscle regeneration and a decrease in fatty infiltration on musculoskeletal magnetic resonance imaging and nine showing improved muscle electrical activity on electromyography. Fifty-three percent of the cases showed an increase in trunk muscle strength, 48% showed an increase in upper limb strength, 59% showed an increase in lower limb strength, and approximately 10% showed improved gait. These data were statistically analyzed using Student's paired t test and found to be significant. The results show that this treatment is safe and efficacious and also improves the quality of life of patients having muscular dystrophy. This manuscript is published as part of the International Association of Neurorestoratology (IANR) supplement issue of Cell Transplantation.
Collapse
Affiliation(s)
- Alok Sharma
- Department of Medical Services and Clinical Research, NeuroGen Brain and Spine Institute, Surana Sethia Hospital and Research Centre, Suman Nagar, Sion-Trombay Road, Chembur, Mumbai, India
| | - Hemangi Sane
- Department of Research & Development, NeuroGen Brain and Spine Institute, Surana Sethia Hospital and Research Centre, Suman Nagar, Sion-Trombay Road, Chembur, Mumbai, India
| | - Prerna Badhe
- Department of Medical Services and Clinical Research, NeuroGen Brain and Spine Institute, Surana Sethia Hospital and Research Centre, Suman Nagar, Sion-Trombay Road, Chembur, Mumbai, India
| | - Nandini Gokulchandran
- Department of Medical Services and Clinical Research, NeuroGen Brain and Spine Institute, Surana Sethia Hospital and Research Centre, Suman Nagar, Sion-Trombay Road, Chembur, Mumbai, India
| | - Pooja Kulkarni
- Department of Research & Development, NeuroGen Brain and Spine Institute, Surana Sethia Hospital and Research Centre, Suman Nagar, Sion-Trombay Road, Chembur, Mumbai, India
| | - Mamta Lohiya
- Department of NeuroRehabilitation, NeuroGen Brain and Spine Institute, Surana Sethia Hospital and Research Centre, Suman Nagar, Sion-Trombay Road, Chembur, Mumbai, India
| | - Hema Biju
- Department of NeuroRehabilitation, NeuroGen Brain and Spine Institute, Surana Sethia Hospital and Research Centre, Suman Nagar, Sion-Trombay Road, Chembur, Mumbai, India
| | - V. C. Jacob
- Department of NeuroRehabilitation, NeuroGen Brain and Spine Institute, Surana Sethia Hospital and Research Centre, Suman Nagar, Sion-Trombay Road, Chembur, Mumbai, India
| |
Collapse
|
92
|
Smythe GM, Forwood JK. Altered mitogen-activated protein kinase signaling in dystrophic (mdx) muscle. Muscle Nerve 2012; 46:374-83. [PMID: 22907228 DOI: 10.1002/mus.23312] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Duchenne muscular dystrophy (DMD) results from a deficiency in the protein, dystrophin. Dystrophic myotubes are susceptible to stressful stimuli. This may be partly due to altered regulation of pro-survival signaling pathways, but a role for mitogen-activated protein (MAP) kinases has not been investigated. METHODS We examined patterns of phosphorylation of key MAP kinase proteins in cultured myotubes responding to oxidative stress, and in muscle tissue in vivo. RESULTS Dystrophic (mdx) myotubes have an increased susceptibility to oxidant-induced death compared with wild-type (C57Bl/10ScSn) myotubes. This correlates with late phosphorylation of c-Jun N-terminal kinase (JNK), and persistently high p38 MAP kinase phosphorylation in mdx myotubes. JNK and extracellular signal-regulated kinase 1/2 (ERK1/2) also showed altered phosphorylation levels in mdx muscle tissue. CONCLUSIONS We show altered patterns of MAP kinase protein phosphorylation in dystrophic muscle in vitro and in vivo. These pathways may be novel pharmacological targets for treating DMD.
Collapse
Affiliation(s)
- Gayle M Smythe
- School of Community Health, Charles Sturt University, P.O. Box 789, Albury, NSW, 2640, Australia.
| | | |
Collapse
|
93
|
Contribution of oxidative stress to pathology in diaphragm and limb muscles with Duchenne muscular dystrophy. J Muscle Res Cell Motil 2012; 34:1-13. [DOI: 10.1007/s10974-012-9330-9] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 10/18/2012] [Indexed: 11/27/2022]
|
94
|
Zolkipli Z, Mai L, Lamhonwah AM, Tein I. The mdx mouse as a model for carnitine deficiency in the pathogenesis of Duchenne muscular dystrophy. Muscle Nerve 2012; 46:767-72. [PMID: 23055315 DOI: 10.1002/mus.23368] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Muscle and cardiac metabolism are dependent on the oxidation of fats and glucose for adenosine triphosphate production, for which L-carnitine is an essential cofactor. METHODS We measured muscle carnitine concentrations in skeletal muscles, diaphragm, and ventricles of C57BL/10ScSn-DMDmdx/J mice (n = 10) and compared them with wild-type C57BL/6J (n = 3), C57BL/10 (n = 10), and C3H (n = 12) mice. Citrate synthase (CS) activity was measured in quadriceps/gluteals and ventricles of mdx and wild-type mice. RESULTS We found significantly lower tissue carnitine in quadriceps/gluteus (P < 0.05) and ventricle (P < 0.05), but not diaphragm of mdx mice, when compared with controls. CS activity was increased in mdx quadriceps/gluteus (P < 0.03) and ventricle (P < 0.02). This suggests compensatory mitochondrial biogenesis. CONCLUSIONS Decreased tissue carnitine has implications for reduced fatty acid and glucose oxidation in mdx quadriceps/gluteus and ventricle. The mdx mouse may be a useful model for studying the role of muscle carnitine deficiency in DMD bioenergetic insufficiency and providing a targeted and timed rationale for L-carnitine therapy.
Collapse
Affiliation(s)
- Zarazuela Zolkipli
- Division of Neurology, Department of Pediatrics, The Hospital for Sick Children, University of Toronto, 555 University Avenue, Toronto, Ontario M5G 1X8, Canada
| | | | | | | |
Collapse
|
95
|
Martins-Bach AB, Bloise AC, Vainzof M, Rahnamaye Rabbani S. Metabolic profile of dystrophic mdx mouse muscles analyzed with in vitro magnetic resonance spectroscopy (MRS). Magn Reson Imaging 2012; 30:1167-76. [DOI: 10.1016/j.mri.2012.04.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 03/13/2012] [Accepted: 04/02/2012] [Indexed: 01/31/2023]
|
96
|
Histone deacetylase inhibitor BML-210 induces growth inhibition and apoptosis and regulates HDAC and DAPC complex expression levels in cervical cancer cells. Mol Biol Rep 2012; 39:10179-86. [PMID: 23007576 DOI: 10.1007/s11033-012-1892-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2012] [Accepted: 09/18/2012] [Indexed: 10/27/2022]
Abstract
Histone deacetylase inhibitors (HDACIs) represent a new class of targeted anti-cancer agents and different other diseases, like muscular disorders. A number of studies have shown that extracellular signal-activated kinases can target chromatin-modifying complexes directly and regulate their function. The molecular connection between the dystrophin-associated protein complex (DAPC) and chromatin has been described, by showing that NO signaling regulates histone deacetylase (HDAC) activity and influences gene expression in different cell types. In present study, we investigated HDACs changes in HeLa cells undergoing growth inhibition and apoptosis, caused by HDACI BML-210 and retinoic acid (ATRA). Cell cycle analysis indicated that HeLa cell treatment with 20 and 30 μM concentration of BML-210 increased the proportion of cells in G0/G1 phase, and caused accumulation in subG1, indicating that the cells are undergoing apoptosis. We determined down-regulation of HDAC 1-5 and 7 after treatment with BML-210. Also, we demonstrated expression of different isoforms of alpha-dystrobrevin (α-DB) and other components of DAPC such as syntrophin, dystrophin, beta-dystrobrevin (β-DB) and NOS in HeLa cells after treatments. We determined changes in protein expression level of dystrophin, NOS1, α- and β-DB and in subcellular localization of α-DB after treatments with BML-210 and ATRA. In conclusion, these results suggest that HDACI BML-210 can inhibit cell growth and induce apoptosis in cervical cancer cells, what correlates with down-regulation of HDAC class I and II and changes in the DAPC expression levels. This can be important for identifying target proteins in DAPC signaling to HDACs, as a target of pharmacological intervention for treatment of muscular dystrophies and other diseases.
Collapse
|
97
|
Fratini F, Macchia G, Torreri P, Matteucci A, Salzano AM, Crescenzi M, Macioce P, Petrucci TC, Ceccarini M. Phosphorylation on threonine 11 of β-dystrobrevin alters its interaction with kinesin heavy chain. FEBS J 2012; 279:4131-44. [PMID: 22978324 DOI: 10.1111/febs.12006] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 09/07/2012] [Accepted: 09/11/2012] [Indexed: 11/30/2022]
Abstract
Dystrobrevin family members (α and β) are cytoplasmic components of the dystrophin-associated glycoprotein complex, a multimeric protein complex first isolated from skeletal muscle, which links the extracellular matrix to the actin cytoskeleton. Dystrobrevin shares high homology with the cysteine-rich and C-terminal domains of dystrophin and a common domain organization. The β-dystrobrevin isoform is restricted to nonmuscle tissues, serves as a scaffold for signaling complexes, and may participate in intracellular transport through its interaction with kinesin heavy chain. We have previously characterized the molecular determinants affecting the β-dystrobrevin-kinesin heavy chain interaction, among which is cAMP-dependent protein kinase [protein kinase A (PKA)] phosphorylation of β-dystrobrevin. In this study, we have identified β-dystrobrevin residues phosphorylated in vitro by PKA with pull-down assays, surface plasmon resonance measurements, and MS analysis. Among the identified phosphorylated residues, we demonstrated, by site-directed mutagenesis, that Thr11 is the regulatory site for the β-dystrobrevin-kinesin interaction. As dystrobrevin may function as a signaling scaffold for kinases/phosphatases, we also investigated whether β-dystrobrevin is phosphorylated in vitro by kinases other than PKA. Thr11 was phosphorylated by protein kinase C, suggesting that this represents a key residue modified by the activation of different signaling pathways.
Collapse
Affiliation(s)
- Federica Fratini
- Department of Cell Biology and Neuroscience, Istituto Superiore di Sanità, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
98
|
Ramírez-Sánchez I, Mendoza-Lorenzo P, Zentella-Dehesa A, Méndez-Bolaina E, Lara-Padilla E, Ceballos-Reyes G, Canto P, Palma-Flores C, Coral-Vázquez RM. Caveolae and non-caveolae lipid raft microdomains of human umbilical vein endothelial cells contain utrophin-associated protein complexes. Biochimie 2012; 94:1884-90. [DOI: 10.1016/j.biochi.2012.05.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2012] [Accepted: 05/01/2012] [Indexed: 12/16/2022]
|
99
|
Juan-Mateu J, Gallano P, Trujillo-Tiebas MJ. Recomendaciones de buena práctica para el diagnóstico genético de las distrofias musculares de Duchenne y de Becker. Med Clin (Barc) 2012; 139:307-12. [DOI: 10.1016/j.medcli.2012.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2011] [Revised: 02/08/2012] [Accepted: 02/09/2012] [Indexed: 01/15/2023]
|
100
|
Moore CJ, Winder SJ. The inside and out of dystroglycan post-translational modification. Neuromuscul Disord 2012; 22:959-65. [PMID: 22770978 DOI: 10.1016/j.nmd.2012.05.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2012] [Revised: 04/27/2012] [Accepted: 05/28/2012] [Indexed: 01/06/2023]
Abstract
In neuromuscular systems dystroglycan provides a vital link between laminin in the extracellular matrix and dystrophin in the membrane cytoskeleton. The integrity of this link is maintained and regulated by post-translational modifications of dystroglycan that have effects both inside and outside the cell. Glycosylation of α-dystroglycan is crucial for its link to laminin and phosphorylation of β-dystroglycan on tyrosine regulates its association with intracellular binding partners. This short review focuses on some of the recent developments in our understanding of the role of these post-translational modification in regulating dystroglycan function, and how new knowledge of signalling through the laminin-dystroglycan axis is leading to hope for treatment for some neuromuscular diseases associated with this adhesion complex.
Collapse
Affiliation(s)
- Chris J Moore
- Department of Biomedical Science, University of Sheffield, Firth Court, Western Bank, Sheffield S10 2TN, UK.
| | | |
Collapse
|