151
|
Terrill JR, Grounds MD, Arthur PG. Increased taurine in pre-weaned juvenile mdx mice greatly reduces the acute onset of myofibre necrosis and dystropathology and prevents inflammation. PLOS CURRENTS 2016; 8:ecurrents.md.77be6ec30e8caf19529a00417614a072. [PMID: 27679740 PMCID: PMC5029885 DOI: 10.1371/currents.md.77be6ec30e8caf19529a00417614a072] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Abstract
BACKGROUND The mdx mouse model for the fatal muscle wasting disease Duchenne Muscular Dystrophy (DMD) shows a very mild pathology once growth has ceased, with low levels of myofibre necrosis in adults. However, from about 3 weeks of post-natal age, muscles of juvenile mdx mice undergo an acute bout of severe necrosis and inflammation: this subsequently decreases and stabilises to lower adult levels by about 6 weeks of age. Prior to the onset of this severe dystropathology, we have shown that mdx mice are deficient in the amino acid taurine (potentially due to weaning), and we propose that this exacerbates myofibre necrosis and inflammation in juvenile mdx mice. OBJECTIVES The purpose of this study was to increase taurine availability to pre-weaned juvenile mdx mice (from 14 days of age), to evaluate the impact on levels of myofibre necrosis and inflammation (at 22 days) during the acute period of severe dystropathology. RESULTS Untreated 22 day old mdx muscle was not deficient in taurine, with similar levels to normal C57 control muscle. However taurine treatment, which increased the taurine content of young dystrophic muscle (by 40%), greatly reduced myofibre necrosis (by 75%) and prevented significant increases in 3 markers of inflammation. CONCLUSION Taurine was very effective at preventing the acute phase of muscle damage that normally results in myofibre necrosis and inflammation in juvenile mdx mice, supporting continued research into the use of taurine as a therapeutic intervention for protecting growing muscles of young DMD boys.
Collapse
Affiliation(s)
| | - Miranda D Grounds
- School of Anatomy and Human Biology, the University of Western Australia, Perth, Australia
| | - Peter G Arthur
- School of Chemistry and Biochemistry, The University of Western Australia, Perth, Western Australia
| |
Collapse
|
152
|
Tjondrokoesoemo A, Schips TG, Sargent MA, Vanhoutte D, Kanisicak O, Prasad V, Lin SCJ, Maillet M, Molkentin JD. Cathepsin S Contributes to the Pathogenesis of Muscular Dystrophy in Mice. J Biol Chem 2016; 291:9920-8. [PMID: 26966179 DOI: 10.1074/jbc.m116.719054] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Indexed: 11/06/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is an X-linked recessive disease caused by mutations in the gene encoding dystrophin. Loss of dystrophin protein compromises the stability of the sarcolemma membrane surrounding each muscle cell fiber, leading to membrane ruptures and leakiness that induces myofiber necrosis, a subsequent inflammatory response, and progressive tissue fibrosis with loss of functional capacity. Cathepsin S (Ctss) is a cysteine protease that is actively secreted in areas of tissue injury and ongoing inflammation, where it participates in extracellular matrix remodeling and healing. Here we show significant induction of Ctss expression and proteolytic activity following acute muscle injury or in muscle from mdx mice, a model of DMD. To examine the functional ramifications associated with greater Ctss expression, the Ctss gene was deleted in the mdx genetic background, resulting in protection from muscular dystrophy pathogenesis that included reduced myofiber turnover and histopathology, reduced fibrosis, and improved running capacity. Mechanistically, deletion of the Ctss gene in the mdx background significantly increased myofiber sarcolemmal membrane stability with greater expression and membrane localization of utrophin, integrins, and β-dystroglycan, which anchor the membrane to the basal lamina and underlying cytoskeletal proteins. Consistent with these results, skeletal muscle-specific transgenic mice overexpressing Ctss showed increased myofiber necrosis, muscle histopathology, and a functional deficit reminiscent of muscular dystrophy. Hence, Ctss induction during muscular dystrophy is a pathologic event that partially underlies disease pathogenesis, and its inhibition might serve as a new therapeutic strategy in DMD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jeffery D Molkentin
- From the Department of Pediatrics and Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, Ohio 45229
| |
Collapse
|
153
|
Griffin DA, Johnson RW, Whitlock JM, Pozsgai ER, Heller KN, Grose WE, Arnold WD, Sahenk Z, Hartzell HC, Rodino-Klapac LR. Defective membrane fusion and repair in Anoctamin5-deficient muscular dystrophy. Hum Mol Genet 2016; 25:1900-1911. [PMID: 26911675 PMCID: PMC5062581 DOI: 10.1093/hmg/ddw063] [Citation(s) in RCA: 82] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 02/22/2016] [Indexed: 11/15/2022] Open
Abstract
Limb-girdle muscular dystrophies are a genetically diverse group of diseases characterized by chronic muscle wasting and weakness. Recessive mutations in ANO5 (TMEM16E) have been directly linked to several clinical phenotypes including limb-girdle muscular dystrophy type 2L and Miyoshi myopathy type 3, although the pathogenic mechanism has remained elusive. ANO5 is a member of the Anoctamin/TMEM16 superfamily that encodes both ion channels and regulators of membrane phospholipid scrambling. The phenotypic overlap of ANO5 myopathies with dysferlin-associated muscular dystrophies has inspired the hypothesis that ANO5, like dysferlin, may be involved in the repair of muscle membranes following injury. Here we show that Ano5-deficient mice have reduced capacity to repair the sarcolemma following laser-induced damage, exhibit delayed regeneration after cardiotoxin injury and suffer from defective myoblast fusion necessary for the proper repair and regeneration of multinucleated myotubes. Together, these data suggest that ANO5 plays an important role in sarcolemmal membrane dynamics. Genbank Mouse Genome Informatics accession no. 3576659.
Collapse
Affiliation(s)
- Danielle A Griffin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital
| | - Ryan W Johnson
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital
| | - Jarred M Whitlock
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Eric R Pozsgai
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, OH, USA and
| | - Kristin N Heller
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital
| | - William E Grose
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital
| | - W David Arnold
- Department of Neurology, Department of Physical Medicine and Rehabilitation, Department of Neuroscience and
| | - Zarife Sahenk
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, Department of Neurology
| | - H Criss Hartzell
- Department of Cell Biology, Emory University School of Medicine, Atlanta, GA, USA
| | - Louise R Rodino-Klapac
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Department of Pediatrics, Integrated Biomedical Science Graduate Program, College of Medicine, The Ohio State University, Columbus, OH, USA and
| |
Collapse
|
154
|
Farini A, Sitzia C, Cassinelli L, Colleoni F, Parolini D, Giovanella U, Maciotta S, Colombo A, Meregalli M, Torrente Y. Inositol 1,4,5-trisphosphate (IP3)-dependent Ca2+ signaling mediates delayed myogenesis in Duchenne muscular dystrophy fetal muscle. Development 2016; 143:658-69. [DOI: 10.1242/dev.126193] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disorder characterized by muscle wasting and premature death. The defective gene is dystrophin, a structural protein, absence of which causes membrane fragility and myofiber necrosis. Several lines of evidence showed that in adult DMD patients dystrophin is involved in signaling pathways that regulate calcium homeostasis and differentiation programs. However, secondary aspects of the disease, such as inflammation and fibrosis development, might represent a bias in the analysis. Because fetal muscle is not influenced by gravity and does not suffer from mechanical load and/or inflammation, we investigated 12-week-old fetal DMD skeletal muscles, highlighting for the first time early alterations in signaling pathways mediated by the absence of dystrophin itself. We found that PLC/IP3/IP3R/Ryr1/Ca2+ signaling is widely active in fetal DMD skeletal muscles and, through the calcium-dependent PKCα protein, exerts a fundamental regulatory role in delaying myogenesis and in myofiber commitment. These data provide new insights into the origin of DMD pathology during muscle development.
Collapse
Affiliation(s)
- Andrea Farini
- Laboratorio di Cellule Staminali, Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Centro Dino Ferrari, Via Francesco Sforza 35, Milan 20122, Centro Dino Ferrari, Italy
| | - Clementina Sitzia
- Laboratorio di Cellule Staminali, Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Centro Dino Ferrari, Via Francesco Sforza 35, Milan 20122, Centro Dino Ferrari, Italy
| | - Letizia Cassinelli
- Laboratorio di Cellule Staminali, Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Centro Dino Ferrari, Via Francesco Sforza 35, Milan 20122, Centro Dino Ferrari, Italy
| | - Federica Colleoni
- Laboratorio di Cellule Staminali, Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Centro Dino Ferrari, Via Francesco Sforza 35, Milan 20122, Centro Dino Ferrari, Italy
| | - Daniele Parolini
- Laboratorio di Cellule Staminali, Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Centro Dino Ferrari, Via Francesco Sforza 35, Milan 20122, Centro Dino Ferrari, Italy
| | - Umberto Giovanella
- Consiglio Nazionale delle Ricerche, Istituto per lo Studio delle Macromolecole (CNR-ISMAC), via Bassini 15, Milano 20133, Italy
| | - Simona Maciotta
- Laboratorio di Cellule Staminali, Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Centro Dino Ferrari, Via Francesco Sforza 35, Milan 20122, Centro Dino Ferrari, Italy
| | - Augusto Colombo
- Servizio ‘Legge 194’ Dipartimento BDN-Fondazione IRCCS, Policlinico Mangiagalli-Regina Elena, Via Francesco Sforza 35, Milan 20122, Italy
| | - Mirella Meregalli
- Laboratorio di Cellule Staminali, Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Centro Dino Ferrari, Via Francesco Sforza 35, Milan 20122, Centro Dino Ferrari, Italy
| | - Yvan Torrente
- Laboratorio di Cellule Staminali, Dipartimento di Fisiopatologia medico-chirurgica e dei Trapianti, Università degli Studi di Milano, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, Milano, Centro Dino Ferrari, Via Francesco Sforza 35, Milan 20122, Centro Dino Ferrari, Italy
| |
Collapse
|
155
|
Terrill JR, Pinniger GJ, Graves JA, Grounds MD, Arthur PG. Increasing taurine intake and taurine synthesis improves skeletal muscle function in the mdx mouse model for Duchenne muscular dystrophy. J Physiol 2016; 594:3095-110. [PMID: 26659826 DOI: 10.1113/jp271418] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 11/18/2015] [Indexed: 11/08/2022] Open
Abstract
KEY POINTS Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease associated with increased inflammation, oxidative stress and myofibre necrosis. Cysteine precursor antioxidants such as N-acetyl cysteine (NAC) and l-2-oxothiazolidine-4-carboxylate (OTC) reduce dystropathology in the mdx mouse model for DMD, and we propose this is via increased synthesis of the amino acid taurine. We compared the capacity of OTC and taurine treatment to increase taurine content of mdx muscle, as well as effects on in vivo and ex vivo muscle function, inflammation and oxidative stress. Both treatments increased taurine in muscles, and improved many aspects of muscle function and reduced inflammation. Taurine treatment also reduced protein thiol oxidation and was overall more effective, as OTC treatment reduced body and muscle weight, suggesting some adverse effects of this drug. These data suggest that increasing dietary taurine is a better candidate for a therapeutic intervention for DMD. ABSTRACT Duchenne muscular dystrophy (DMD) is a fatal muscle wasting disease for which there is no widely available cure. Whilst the mechanism of loss of muscle function in DMD and the mdx mouse model are not fully understood, disruptions in intracellular calcium homeostasis, inflammation and oxidative stress are implicated. We have shown that protein thiol oxidation is increased in mdx muscle, and that the indirect thiol antioxidant l-2-oxothiazolidine-4-carboxylate (OTC), which increases cysteine availability, decreases pathology and increases in vivo strength. We propose that the protective effects of OTC are a consequence of conversion of cysteine to taurine, which has itself been shown to be beneficial to mdx pathology. This study compares the efficacy of taurine with OTC in decreasing dystropathology in mdx mice by measuring in vivo and ex vivo contractile function and measurements of inflammation and protein thiol oxidation. Increasing the taurine content of mdx muscle improved both in vivo and ex vivo muscle strength and function, potentially via anti-inflammatory and antioxidant effects of taurine. OTC treatment increased taurine synthesis in the liver and taurine content of mdx muscle, improved muscle function and decreased inflammation. However, OTC was less effective than taurine treatment, with OTC also decreasing body and EDL muscle weights, suggesting that OTC had some detrimental effects. These data support continued research into the use of taurine as a therapeutic intervention for DMD, and suggest that increasing dietary taurine is the better strategy for increasing taurine content and decreasing severity of dystropathology.
Collapse
Affiliation(s)
- Jessica R Terrill
- School of Chemistry and Biochemistry, the University of Western Australia, Perth, Western Australia.,School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, Western Australia
| | - Gavin J Pinniger
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, Western Australia
| | - Jamie A Graves
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, Western Australia
| | - Miranda D Grounds
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, Western Australia
| | - Peter G Arthur
- School of Chemistry and Biochemistry, the University of Western Australia, Perth, Western Australia
| |
Collapse
|
156
|
Tjondrokoesoemo A, Schips T, Kanisicak O, Sargent MA, Molkentin JD. Genetic overexpression of Serpina3n attenuates muscular dystrophy in mice. Hum Mol Genet 2016; 25:1192-202. [PMID: 26744329 DOI: 10.1093/hmg/ddw005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 01/05/2016] [Indexed: 01/06/2023] Open
Abstract
Muscular dystrophy (MD) is associated with mutations in genes that stabilize the myofiber plasma membrane, such as through the dystrophin-glycoprotein complex (DGC). Instability of this complex or defects in membrane repair/integrity leads to calcium influx and myofiber necrosis leading to progressive dystrophic disease. MD pathogenesis is also associated with increased skeletal muscle protease levels and activity that could augment weakening of the sarcolemma through greater degradation of cellular attachment complexes. Here, we observed a compensatory increase in the serine protease inhibitor Serpina3n in mouse models of MD and after acute muscle tissue injury. Serpina3n muscle-specific transgenic mice were generated to model this increase in expression, which reduced the activity of select proteases in dystrophic skeletal muscle and protected muscle from both acute injury with cardiotoxin and from chronic muscle disease in the mdx or Sgcd(-/-) MD genetic backgrounds. The Serpina3n transgene mitigated muscle degeneration and fibrosis, reduced creatine kinase serum levels, restored running capacity on a treadmill and reduced muscle membrane leakiness in vivo that is characteristic of mdx and Sgcd(-/-) mice. Mechanistically, we show that increased Serpina3n promotes greater sarcolemma membrane integrity and stability in dystrophic mouse models in association with increased membrane residence of the integrins, the DGC/utrophin-glycoprotein complex of proteins and annexin A1. Hence, Serpina3n blocks endogenous increases in the activity of select skeletal muscle resident proteases during injury or dystrophic disease, which stabilizes the sarcolemma leading to less myofiber degeneration and increased regeneration. These results suggest the use of select protease inhibitors as a strategy for treating MD.
Collapse
Affiliation(s)
| | - Tobias Schips
- Department of Pediatrics, University of Cincinnati and
| | | | | | - Jeffery D Molkentin
- Department of Pediatrics, University of Cincinnati and Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, 240 Albert Sabin Way, MLC7020, Cincinnati, OH 45229, USA
| |
Collapse
|
157
|
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: 207] [Impact Index Per Article: 23.0] [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
|
158
|
A Haplotype of Two Novel Polymorphisms in δ-Sarcoglycan Gene Increases Risk of Dilated Cardiomyopathy in Mongoloid Population. PLoS One 2015; 10:e0145602. [PMID: 26720722 PMCID: PMC4697846 DOI: 10.1371/journal.pone.0145602] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 12/07/2015] [Indexed: 11/21/2022] Open
Abstract
The role of genetic abnormality of δ-sarcoglycan (δ-SG) gene in dilated (DCM) and hypertrophied (HCM) cardiomyopathy patients is still unfolding. In this study we first defined the promoter region and then searched for polymorphisms/mutations among the promoter, 5'-untranslated region, and the encoding exons in δ-SG gene in 104 Chinese patients with DCM, 145 with HCM, and 790 normal controls. Two novel polymorphisms were found, an 11 base-pair (bp) deletion (c.-100~-110; -) in the promoter region and a missense polymorphism of A848G resulting in p.Q283R in the highly conserved C-terminus. The prevalence of homozygous genotype -/- of c.-100~-110 was slightly higher in DCM (14.42%) and HCM patients (14.48%), as compared with normal controls (11.01%). The prevalence of genotype of 848A/G was significantly higher in DCM (6.73%; OR = 9.43; p = 0.0002), but not in HCM patients (1.38%; OR = 1.37; p = 0.62), as compared with controls (0.76%). Haplotype -_G consisting c.-100~-110 and A848G was associated with increased risk of DCM (OR = 17.27; 95%CI = 3.19–93.56; p = 0.001) but not associated with HCM (OR = 1.90; 95%CI = 0.38–9.55; p = 0.44). Co-occurrence of the genotypes -/- of c.-100~-110 and 848A/G was found in 5 patients with DCM (4.81%; OR = 39.85; p = 0.0001), none of HCM patients, and only 1 of the controls (0.13%). Both polymorphisms were also found in the Japanese population, but not in the Africans and Caucasians. C.-100~-110 resulted in a decrease of δ-SG promoter activity to 64±3% of the control level (p<0.01). Both co-immunoprecipitation and in vitro protein pull-down assays demonstrated that δ-SG-283R interacts normally to β- and γ-SG, but significantly decreased localization of β/δ/γ-SG on the plasma membrane. In conclusion, haplotype -_G composed of c.-100~-110 and A848G confers higher susceptibility to DCM in the Mongoloid population.
Collapse
|
159
|
Zhang L, Liu Z, Hu KY, Tian QB, Wei LG, Zhao Z, Shen HR, Hu J. Early myocardial damage assessment in dystrophinopathies using (99)Tc(m)-MIBI gated myocardial perfusion imaging. Ther Clin Risk Manag 2015; 11:1819-27. [PMID: 26677332 PMCID: PMC4677759 DOI: 10.2147/tcrm.s89962] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Background Early detection of muscular dystrophy (MD)-associated cardiomyopathy is important because early medical treatment may slow cardiac remodeling and attenuate symptoms of cardiac dysfunction; however, no sensitive and standard diagnostic method for MD at an earlier stage has been well-recognized. Thus, the aim of this study was to test the early diagnostic value of technetium 99m-methoxyisobutylisonitrile (99Tcm-MIBI) gated myocardial perfusion imaging (G-MPI) for MD. Methods and results Ninety-one patients underwent 99Tcm-MIBI G-MPI examinations when they were diagnosed with Duchenne muscular dystrophy (DMD) (n=77) or Becker muscular dystrophy (BMD; n=14). 99Tcm-MIBI G-MPI examinations were repeated in 43 DMD patients who received steroid treatments for 2 years as a follow-up examination. Myocardial defects were observed in nearly every segment of the left ventricular wall in both DMD and BMD patients compared with controls, especially in the inferior walls and the apices by using 99Tcm-MIBI G-MPI. Cardiac wall movement impairment significantly correlated with age in the DMD and BMD groups (rs=0.534 [P<0.05] and rs=0.784 [P<0.05], respectively). Intermittent intravenous doses of glucocorticoids and continuation with oral steroid treatments significantly improved myocardial function in DMD patients (P<0.05), but not in BMD patients. Conclusion 99Tcm-MIBI G-MPI is a sensitive and safe approach for early evaluation of cardiomyopathy in patients with DMD or BMD, and can serve as a candidate method for the evaluation of progression, prognosis, and assessment of the effect of glucocorticoid treatment in these patients.
Collapse
Affiliation(s)
- Li Zhang
- Department of Cardiovascular Disorders
| | - Zhe Liu
- Department of Geriatrics, The Third Hospital of Hebei Medical University
| | - Ke-You Hu
- The Public Health Department, Hebei Medical University
| | - Qing-Bao Tian
- The Public Health Department, Hebei Medical University
| | | | - Zhe Zhao
- Department of Neuromuscular Disorders, The Third Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Hong-Rui Shen
- Department of Neuromuscular Disorders, The Third Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| | - Jing Hu
- Department of Neuromuscular Disorders, The Third Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
| |
Collapse
|
160
|
Smith SJ, Horstick EJ, Davidson AE, Dowling J. Analysis of Zebrafish Larvae Skeletal Muscle Integrity with Evans Blue Dye. J Vis Exp 2015:53183. [PMID: 26649573 PMCID: PMC4692762 DOI: 10.3791/53183] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The zebrafish model is an emerging system for the study of neuromuscular disorders. In the study of neuromuscular diseases, the integrity of the muscle membrane is a critical disease determinant. To date, numerous neuromuscular conditions display degenerating muscle fibers with abnormal membrane integrity; this is most commonly observed in muscular dystrophies. Evans Blue Dye (EBD) is a vital, cell permeable dye that is rapidly taken into degenerating, damaged, or apoptotic cells; in contrast, it is not taken up by cells with an intact membrane. EBD injection is commonly employed to ascertain muscle integrity in mouse models of neuromuscular diseases. However, such EBD experiments require muscle dissection and/or sectioning prior to analysis. In contrast, EBD uptake in zebrafish is visualized in live, intact preparations. Here, we demonstrate a simple and straightforward methodology for performing EBD injections and analysis in live zebrafish. In addition, we demonstrate a co-injection strategy to increase efficacy of EBD analysis. Overall, this video article provides an outline to perform EBD injection and characterization in zebrafish models of neuromuscular disease.
Collapse
Affiliation(s)
- Sarah J Smith
- Program in Genetics & Genome Biology, The Hospital for Sick Children; Department of Molecular Genetics, The University of Toronto
| | - Eric J Horstick
- Program in Genomics of Differentiation, Eunice Kennedy Shriver National Institute of Child Health and Human Development; Departments of Pediatrics and Neurology, University of Michigan
| | - Ann E Davidson
- Program in Genetics & Genome Biology, The Hospital for Sick Children; Department of Molecular Genetics, The University of Toronto
| | - James Dowling
- Program in Genetics & Genome Biology, The Hospital for Sick Children; Department of Molecular Genetics, The University of Toronto; Departments of Pediatrics and Neurology, University of Michigan;
| |
Collapse
|
161
|
Crosstalk between RyR2 oxidation and phosphorylation contributes to cardiac dysfunction in mice with Duchenne muscular dystrophy. J Mol Cell Cardiol 2015; 89:177-84. [PMID: 26555638 DOI: 10.1016/j.yjmcc.2015.11.009] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/12/2015] [Revised: 10/28/2015] [Accepted: 11/05/2015] [Indexed: 12/24/2022]
Abstract
BACKGROUND Patients with Duchenne muscular dystrophy (DMD) are at risk of developing cardiomyopathy and cardiac arrhythmias. Studies in a mouse model of DMD revealed that enhanced sarcoplasmic reticulum (SR) Ca(2+) leak contributes to the pathogenesis of cardiac dysfunction. In view of recent data suggesting the involvement of altered phosphorylation and oxidation of the cardiac ryanodine receptor (RyR2)/Ca(2+) release channel, we hypothesized that inhibition of RyR2 phosphorylation in a mouse model of DMD can prevent SR Ca(2+) leak by reducing RyR2 oxidation. METHODS AND RESULTS Confocal Ca(2+) imaging and single RyR2 channel recordings revealed that both inhibition of S2808 or S2814 phosphorylation, and inhibition of oxidation could normalize RyR2 activity in mdx mice. Moreover, Western blotting revealed that genetic inhibition of RyR2 phosphorylation at S2808 or S2814 reduced RyR2 oxidation. Production of reactive oxygen species (ROS) in myocytes from mdx mice was reduced by both inhibition of RyR2 phosphorylation or the ROS scavenger 2-mercaptopropionyl glycine (MPG). Finally, it was shown that ROS production in mdx mice is proportional to the activity of RyR2-mediated SR Ca(2+) leak, and likely generated by Nox2. CONCLUSIONS Increased ROS production in the hearts of mdx mice drives the progression of cardiac dysfunction. Inhibition of RyR2 phosphorylation can suppress SR Ca(2+) leak in mdx mouse hearts in part by reducing RyR2 oxidation.
Collapse
|
162
|
Barzilai-Tutsch H, Bodanovsky A, Maimon H, Pines M, Halevy O. Halofuginone promotes satellite cell activation and survival in muscular dystrophies. Biochim Biophys Acta Mol Basis Dis 2015; 1862:1-11. [PMID: 26454207 DOI: 10.1016/j.bbadis.2015.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2015] [Revised: 10/05/2015] [Accepted: 10/06/2015] [Indexed: 11/18/2022]
Abstract
Halofuginone is a leading agent in preventing fibrosis and inflammation in various muscular dystrophies. We hypothesized that in addition to these actions, halofuginone directly promotes the cell-cycle events of satellite cells in the mdx and dysf(-/-) mouse models of early-onset Duchenne muscular dystrophy and late-onset dysferlinopathy, respectively. In both models, addition of halofuginone to freshly prepared single gastrocnemius myofibers derived from 6-week-old mice increased BrdU incorporation at as early as 18h of incubation, as well as phospho-histone H3 (PHH3) and MyoD protein expression in the attached satellite cells, while having no apparent effect on myofibers derived from wild-type mice. BrdU incorporation was abolished by an inhibitor of mitogen-activated protein kinase/extracellular signal-regulated protein kinase, suggesting involvement of this pathway in mediating halofuginone's effects on cell-cycle events. In cultures of myofibers and myoblasts isolated from dysf(-/-) mice, halofuginone reduced Bax and induced Bcl2 expression levels and induced Akt phosphorylation in a time-dependent manner. Addition of an inhibitor of the phosphinositide-3-kinase/Akt pathway reversed the halofuginone-induced cell survival, suggesting this pathway's involvement in mediating halofuginone's effects on survival. Thus, in addition to its known role in inhibiting fibrosis and inflammation, halofuginone plays a direct role in satellite cell activity and survival in muscular dystrophies, regardless of the mutation. These actions are of the utmost importance for improving muscle pathology and function in muscular dystrophies.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Cell Cycle/drug effects
- Cell Survival/drug effects
- MAP Kinase Signaling System/drug effects
- Male
- Mice
- Mice, Inbred C57BL
- Muscle Fibers, Skeletal/cytology
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscular Dystrophies, Limb-Girdle/drug therapy
- Muscular Dystrophies, Limb-Girdle/metabolism
- Muscular Dystrophies, Limb-Girdle/pathology
- Muscular Dystrophy, Duchenne/drug therapy
- Muscular Dystrophy, Duchenne/metabolism
- Muscular Dystrophy, Duchenne/pathology
- Phosphatidylinositol 3-Kinases/metabolism
- Piperidines/pharmacology
- Piperidines/therapeutic use
- Proto-Oncogene Proteins c-akt/metabolism
- Quinazolinones/pharmacology
- Quinazolinones/therapeutic use
- Satellite Cells, Skeletal Muscle/cytology
- Satellite Cells, Skeletal Muscle/drug effects
- Satellite Cells, Skeletal Muscle/metabolism
- Satellite Cells, Skeletal Muscle/pathology
- Signal Transduction/drug effects
Collapse
Affiliation(s)
- Hila Barzilai-Tutsch
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Anna Bodanovsky
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Hadar Maimon
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel
| | - Mark Pines
- Institute of Animal Science, The Volcani Center, Bet Dagan 52505, Israel
| | - Orna Halevy
- Department of Animal Sciences, The Hebrew University of Jerusalem, Rehovot 76100, Israel.
| |
Collapse
|
163
|
Gonzalez JP, Ramachandran J, Xie LH, Contreras JE, Fraidenraich D. Selective Connexin43 Inhibition Prevents Isoproterenol-Induced Arrhythmias and Lethality in Muscular Dystrophy Mice. Sci Rep 2015; 5:13490. [PMID: 26311238 PMCID: PMC4550874 DOI: 10.1038/srep13490] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2015] [Accepted: 07/27/2015] [Indexed: 12/18/2022] Open
Abstract
Duchenne muscular dystrophy (DMD) is caused by an X-linked mutation that leads to the absence of dystrophin, resulting in life-threatening arrhythmogenesis and associated heart failure. We targeted the gap junction protein connexin43 (Cx43) responsible for maintaining cardiac conduction. In mild mdx and severe mdx:utr mouse models of DMD, and human DMD tissues, Cx43 was found to be pathologically mislocalized to lateral sides of cardiomyocytes. In addition, overall Cx43 protein levels were markedly increased in mouse and human DMD heart tissues examined. Electrocardiography on isoproterenol challenged mice showed that both models developed arrhythmias and died within 24 hours, while wild-type mice were free of pathology. Administering peptide mimetics to inhibit lateralized Cx43 function prior to challenge protected mdx mice from arrhythmogenesis and death, while mdx:utr mice displayed markedly improved ECG scores. These findings suggest that Cx43 lateralization contributes significantly to DMD arrhythmogenesis and that selective inhibition may provide substantial benefit.
Collapse
Affiliation(s)
- J Patrick Gonzalez
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ USA
| | - Jayalakshmi Ramachandran
- Department of Pharmacology and Physiology, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ USA
| | - Lai-Hua Xie
- Department of Pharmacology and Physiology, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ USA
| | - Jorge E Contreras
- Department of Pharmacology and Physiology, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ USA
| | - Diego Fraidenraich
- Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, New Jersey Medical School, Newark, NJ USA
| |
Collapse
|
164
|
Terrill JR, Grounds MD, Arthur PG. Taurine deficiency, synthesis and transport in the mdx mouse model for Duchenne Muscular Dystrophy. Int J Biochem Cell Biol 2015; 66:141-8. [PMID: 26239309 DOI: 10.1016/j.biocel.2015.07.016] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/02/2015] [Accepted: 07/29/2015] [Indexed: 02/06/2023]
Abstract
The amino acid taurine is essential for the function of skeletal muscle and administration is proposed as a treatment for Duchenne Muscular Dystrophy (DMD). Taurine homeostasis is dependent on multiple processes including absorption of taurine from food, endogenous synthesis from cysteine and reabsorption in the kidney. This study investigates the cause of reported taurine deficiency in the dystrophic mdx mouse model of DMD. Levels of metabolites (taurine, cysteine, cysteine sulfinate and hypotaurine) and proteins (taurine transporter [TauT], cysteine deoxygenase and cysteine sulfinate dehydrogenase) were quantified in juvenile control C57 and dystrophic mdx mice aged 18 days, 4 and 6 weeks. In C57 mice, taurine content was much higher in both liver and plasma at 18 days, and both cysteine and cysteine deoxygenase were increased. As taurine levels decreased in maturing C57 mice, there was increased transport (reabsorption) of taurine in the kidney and muscle. In mdx mice, taurine and cysteine levels were much lower in liver and plasma at 18 days, and in muscle cysteine was low at 18 days, whereas taurine was lower at 4: these changes were associated with perturbations in taurine transport in liver, kidney and muscle and altered metabolism in liver and kidney. These data suggest that the maintenance of adequate body taurine relies on sufficient dietary intake of taurine and cysteine availability and metabolism, as well as retention of taurine by the kidney. This research indicates dystrophin deficiency not only perturbs taurine metabolism in the muscle but also affects taurine metabolism in the liver and kidney, and supports targeting cysteine and taurine deficiency as a potential therapy for DMD.
Collapse
Affiliation(s)
- Jessica R Terrill
- School of Chemistry and Biochemistry, University of Western Australia, Crawley, WA, Australia; School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, WA, Australia.
| | - Miranda D Grounds
- School of Anatomy, Physiology and Human Biology, the University of Western Australia, Perth, WA, Australia
| | - Peter G Arthur
- School of Chemistry and Biochemistry, University of Western Australia, Crawley, WA, Australia
| |
Collapse
|
165
|
Kimura A. Molecular genetics and pathogenesis of cardiomyopathy. J Hum Genet 2015; 61:41-50. [PMID: 26178429 DOI: 10.1038/jhg.2015.83] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 06/15/2015] [Indexed: 12/19/2022]
Abstract
Cardiomyopathy is defined as a disease of functional impairment in the cardiac muscle and its etiology includes both extrinsic and intrinsic factors. Cardiomyopathy caused by the intrinsic factors is called as primary cardiomyopathy of which two major clinical phenotypes are hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Genetic approaches have revealed the disease genes for hereditary primary cardiomyopathy and functional studies have demonstrated that characteristic functional alterations induced by the disease-associated mutations are closely related to the clinical types, such that increased and decreased Ca(2+) sensitivities of muscle contraction are associated with HCM and DCM, respectively. In addition, recent studies have suggested that mutations in the Z-disc components found in HCM and DCM may result in increased and decreased stiffness of sarcomere, respectively. Moreover, functional analysis of mutations in the other components of cardiac muscle have suggested that the altered response to metabolic stresses is associated with cardiomyopathy, further indicating the heterogeneity in the etiology and pathogenesis of cardiomyopathy.
Collapse
Affiliation(s)
- Akinori Kimura
- Department of Molecular Pathogenesis, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
166
|
Echarri A, Del Pozo MA. Caveolae - mechanosensitive membrane invaginations linked to actin filaments. J Cell Sci 2015; 128:2747-58. [PMID: 26159735 DOI: 10.1242/jcs.153940] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
An essential property of the plasma membrane of mammalian cells is its plasticity, which is required for sensing and transmitting of signals, and for accommodating the tensional changes imposed by its environment or its own biomechanics. Caveolae are unique invaginated membrane nanodomains that play a major role in organizing signaling, lipid homeostasis and adaptation to membrane tension. Caveolae are frequently associated with stress fibers, a major regulator of membrane tension and cell shape. In this Commentary, we discuss recent studies that have provided new insights into the function of caveolae and have shown that trafficking and organization of caveolae are tightly regulated by stress-fiber regulators, providing a functional link between caveolae and stress fibers. Furthermore, the tension in the plasma membrane determines the curvature of caveolae because they flatten at high tension and invaginate at low tension, thus providing a tension-buffering system. Caveolae also regulate multiple cellular pathways, including RhoA-driven actomyosin contractility and other mechanosensitive pathways, suggesting that caveolae could couple mechanotransduction pathways to actin-controlled changes in tension through their association with stress fibers. Therefore, we argue here that the association of caveolae with stress fibers could provide an important strategy for cells to deal with mechanical stress.
Collapse
Affiliation(s)
- Asier Echarri
- Integrin Signaling Laboratory, Cell Biology & Physiology Program, Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| | - Miguel A Del Pozo
- Integrin Signaling Laboratory, Cell Biology & Physiology Program, Cell & Developmental Biology Area, Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Melchor Fernández Almagro, 3, Madrid 28029, Spain
| |
Collapse
|
167
|
Sienkiewicz D, Kulak W, Okurowska-Zawada B, Paszko-Patej G, Kawnik K. Duchenne muscular dystrophy: current cell therapies. Ther Adv Neurol Disord 2015; 8:166-77. [PMID: 26136844 PMCID: PMC4480531 DOI: 10.1177/1756285615586123] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Duchenne muscular dystrophy is a genetically determined X-linked disease and the most common, progressive pediatric muscle disorder. For decades, research has been conducted to find an effective therapy. This review presents current therapeutic methods for Duchenne muscular dystrophy, based on scientific articles in English published mainly in the period 2000 to 2014. We used the PubMed database to identify and review the most important studies. An analysis of contemporary studies of stem cell therapy and the use of granulocyte colony-stimulating factor (G-CSF) in muscular dystrophy was performed.
Collapse
Affiliation(s)
- Dorota Sienkiewicz
- Department of Pediatric Rehabilitation Medical University, Bialystok, Poland
| | - Wojciech Kulak
- Department of Pediatric Rehabilitation, Medical University of Białystok, 15-274 Bialystok, 17 Waszyngtona street, Poland
| | | | | | - Katarzyna Kawnik
- Department of Pediatric Rehabilitation Medical University, Bialystok, Poland
| |
Collapse
|
168
|
Wu H, Lee J, Vincent LG, Wang Q, Gu M, Lan F, Churko JM, Sallam KI, Matsa E, Sharma A, Gold JD, Engler AJ, Xiang YK, Bers DM, Wu JC. Epigenetic Regulation of Phosphodiesterases 2A and 3A Underlies Compromised β-Adrenergic Signaling in an iPSC Model of Dilated Cardiomyopathy. Cell Stem Cell 2015; 17:89-100. [PMID: 26095046 DOI: 10.1016/j.stem.2015.04.020] [Citation(s) in RCA: 139] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 03/01/2015] [Accepted: 04/28/2015] [Indexed: 02/07/2023]
Abstract
β-adrenergic signaling pathways mediate key aspects of cardiac function. Its dysregulation is associated with a range of cardiac diseases, including dilated cardiomyopathy (DCM). Previously, we established an iPSC model of familial DCM from patients with a mutation in TNNT2, a sarcomeric protein. Here, we found that the β-adrenergic agonist isoproterenol induced mature β-adrenergic signaling in iPSC-derived cardiomyocytes (iPSC-CMs) but that this pathway was blunted in DCM iPSC-CMs. Although expression levels of several β-adrenergic signaling components were unaltered between control and DCM iPSC-CMs, we found that phosphodiesterases (PDEs) 2A and PDE3A were upregulated in DCM iPSC-CMs and that PDE2A was also upregulated in DCM patient tissue. We further discovered increased nuclear localization of mutant TNNT2 and epigenetic modifications of PDE genes in both DCM iPSC-CMs and patient tissue. Notably, pharmacologic inhibition of PDE2A and PDE3A restored cAMP levels and ameliorated the impaired β-adrenergic signaling of DCM iPSC-CMs, suggesting therapeutic potential.
Collapse
Affiliation(s)
- Haodi Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jaecheol Lee
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ludovic G Vincent
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Qingtong Wang
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Mingxia Gu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Feng Lan
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jared M Churko
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Karim I Sallam
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elena Matsa
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Arun Sharma
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joseph D Gold
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Adam J Engler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA; Sanford Consortium for Regenerative Medicine, La Jolla, CA 92037, USA
| | - Yang K Xiang
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Donald M Bers
- Department of Pharmacology, University of California, Davis, Davis, CA 95616, USA
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Division of Cardiology, Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
169
|
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; 36:155-167. [PMID: 25669899 DOI: 10.1007/s10974-015-9406-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
170
|
Petri H, Sveen ML, Thune JJ, Vissing C, Dahlqvist JR, Witting N, Bundgaard H, Køber L, Vissing J. Progression of cardiac involvement in patients with limb-girdle type 2 and Becker muscular dystrophies: A 9-year follow-up study. Int J Cardiol 2015; 182:403-11. [DOI: 10.1016/j.ijcard.2014.12.090] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2014] [Revised: 12/02/2014] [Accepted: 12/25/2014] [Indexed: 01/22/2023]
|
171
|
Stephens NA, Skipworth RJE, Gallagher IJ, Greig CA, Guttridge DC, Ross JA, Fearon KCH. Evaluating potential biomarkers of cachexia and survival in skeletal muscle of upper gastrointestinal cancer patients. J Cachexia Sarcopenia Muscle 2015; 6:53-61. [PMID: 26136412 PMCID: PMC4435097 DOI: 10.1002/jcsm.12005] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Revised: 08/18/2014] [Accepted: 09/10/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND In order to grow the potential therapeutic armamentarium in the cachexia domain of supportive oncology, there is a pressing need to develop suitable biomarkers and potential drug targets. This pilot study evaluated several potential candidate biomarkers in skeletal muscle biopsies from a cohort of upper gastrointestinal cancer (UGIC) patients. METHODS One hundred seven patients (15 weight-stable healthy controls (HC) and 92 UGIC patients) were recruited. Mean (standard deviation) weight-loss of UGIC patients was 8.1 (9.3%). Cachexia was defined as weight-loss ≥5%. Rectus abdominis muscle was obtained at surgery and was analysed by western blotting or quantitative real-time-polymerase chain reaction. Candidate markers were selected according to previous literature and included Akt and phosphorylated Akt (pAkt, n = 52), forkhead box O transcription factors (n = 59), ubiquitin E3 ligases (n = 59, control of muscle anabolism/catabolism), BNIP3 and GABARAPL1 (n = 59, as markers of autophagy), myosin heavy-chain (MyHC, n = 54), dystrophin (n = 39), β-dystroglycan (n = 52), and β-sarcoglycan (n = 52, as markers of structural alteration in a muscle). Patients were followed up for an average of 1255 days (range 581-1955 days) or until death. Patients were grouped accordingly and analysed by (i) all cancer patients vs. HC; (ii) cachectic vs. non-cachectic cancer patients; and (iii) cancer patients surviving ≤1 vs. >1 year post operatively. RESULTS Cancer compared with HC patients had reduced mean (standard deviation) total Akt protein [0.49 (0.31) vs. 0.89 (0.17), P = 0.001], increased ratio of phosphorylated to total Akt [1.33 (1.04) vs. 0.32 (0.21), P = 0.002] and increased expression of GABARAPL1 [1.60 (0.76) vs. 1.10 (0.57), P = 0.024]. β-Dystroglycan levels were higher in cachectic compared with non-cachectic cancer patients [1.01 (0.16) vs. 0.87 (0.20), P = 0.007]. Survival was shortened in patients with low compared with high MyHC levels (median 316 vs. 1326 days, P = 0.023) and dystrophin levels (median 341 vs. 660 days, P = 0.008). CONCLUSIONS The present study has identified intramuscular protein level of β-dystroglycan as a potential biomarker of cancer cachexia. Changes in the structural elements of muscle (MyHC or dystrophin) appear to be survival biomarkers.
Collapse
Affiliation(s)
- Nathan A Stephens
- Department of Clinical and Surgical Sciences (Surgery), School of Clinical Sciences, University of Edinburgh, Royal Infirmary, 51 Little France Crescent, Edinburgh, EH16 4SA, UK
| | - Richard J E Skipworth
- Department of Clinical and Surgical Sciences (Surgery), School of Clinical Sciences, University of Edinburgh, Royal Infirmary, 51 Little France Crescent, Edinburgh, EH16 4SA, UK
| | - Iain J Gallagher
- Department of Clinical and Surgical Sciences (Surgery), School of Clinical Sciences, University of Edinburgh, Royal Infirmary, 51 Little France Crescent, Edinburgh, EH16 4SA, UK
| | - Carolyn A Greig
- Department of Clinical and Surgical Sciences (Surgery), School of Clinical Sciences, University of Edinburgh, Royal Infirmary, 51 Little France Crescent, Edinburgh, EH16 4SA, UK
| | - Denis C Guttridge
- Division of Human Cancer Genetics, The Ohio State University Wexner Medical Center, 410 W. 10th Ave., Columbus, OH, 43210, USA
| | - James A Ross
- Department of Clinical and Surgical Sciences (Surgery), School of Clinical Sciences, University of Edinburgh, Royal Infirmary, 51 Little France Crescent, Edinburgh, EH16 4SA, UK
| | - Kenneth C H Fearon
- Department of Clinical and Surgical Sciences (Surgery), School of Clinical Sciences, University of Edinburgh, Royal Infirmary, 51 Little France Crescent, Edinburgh, EH16 4SA, UK
| |
Collapse
|
172
|
Spinazzola JM, Smith TC, Liu M, Luna EJ, Barton ER. Gamma-sarcoglycan is required for the response of archvillin to mechanical stimulation in skeletal muscle. Hum Mol Genet 2015; 24:2470-81. [PMID: 25605665 DOI: 10.1093/hmg/ddv008] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2014] [Accepted: 01/13/2015] [Indexed: 01/23/2023] Open
Abstract
Loss of gamma-sarcoglycan (γ-SG) induces muscle degeneration and signaling defects in response to mechanical load, and its absence is common to both Duchenne and limb girdle muscular dystrophies. Growing evidence suggests that aberrant signaling contributes to the disease pathology; however, the mechanisms of γ-SG-mediated mechanical signaling are poorly understood. To uncover γ-SG signaling pathway components, we performed yeast two-hybrid screens and identified the muscle-specific protein archvillin as a γ-SG and dystrophin interacting protein. Archvillin protein and message levels were significantly upregulated at the sarcolemma of murine γ-SG-null (gsg(-/-)) muscle but delocalized in dystrophin-deficient mdx muscle. Similar elevation of archvillin protein was observed in human quadriceps muscle lacking γ-SG. Reintroduction of γ-SG in gsg(-/-) muscle by rAAV injection restored archvillin levels to that of control C57 muscle. In situ eccentric contraction of tibialis anterior (TA) muscles from C57 mice caused ERK1/2 phosphorylation, nuclear activation of P-ERK1/2 and stimulus-dependent archvillin association with P-ERK1/2. In contrast, TA muscles from gsg(-/-) and mdx mice exhibited heightened P-ERK1/2 and increased nuclear P-ERK1/2 localization following eccentric contractions, but the archvillin-P-ERK1/2 association was completely ablated. These results position archvillin as a mechanically sensitive component of the dystrophin complex and demonstrate that signaling defects caused by loss of γ-SG occur both at the sarcolemma and in the nucleus.
Collapse
Affiliation(s)
- Janelle M Spinazzola
- Department of Anatomy and Cell Biology, School of Dental Medicine, Pennsylvania Muscle Institute, and
| | - Tara C Smith
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Min Liu
- Department of Physiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA and
| | - Elizabeth J Luna
- Department of Cell and Developmental Biology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Elisabeth R Barton
- Department of Anatomy and Cell Biology, School of Dental Medicine, Pennsylvania Muscle Institute, and
| |
Collapse
|
173
|
O'Brien L, Varadi R, Goldstein RS, Evans RA. Cardiac management of ventilator-assisted individuals with Duchenne muscular dystrophy. Chron Respir Dis 2015; 11:103-10. [PMID: 24728656 DOI: 10.1177/1479972314529673] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
As life expectancy of patients with Duchenne muscular dystrophy (DMD) has increased to the 5th decade, in part due to improved ventilatory support, cardiomyopathy is projected to increase as a cause of death. International guidelines recommend an annual assessment of cardiac function and initiation of appropriate pharmacological treatment. We conducted an audit of the cardiac management in patients with DMD requiring ventilatory support and reported a case series of the collated cardiac investigations. Patients with DMD requiring ventilatory support were included in the study. The date of the last electrocardiogram (ECG), echocardiogram (ECHO), cardiology review and pharmacological management were retrieved from the medical records. If an annual cardiac assessment had not been performed this was requested and the latest ECGs and ECHO reports were collated. A total of 30 patients with DMD (29 males, mean (SD) age of 30 (7) years) met the inclusion criteria. Although there was ECG and ECHO documentation in 24 and 21 individuals, respectively, it was only recent in 10 and 6 individuals. In all, 60% of patients had been assessed by a cardiologist, but only 10% within the last year. Over half of the patients failed to attend their new appointments. From the available results, 18 of the 19 patients had an abnormal ECG, 11 of the 16 patients had left ventricular (LV) impairment and 55% of patients had a change in prescription following cardiac investigations. There is a need for a coordinated cardiorespiratory approach towards adult patients with DMD. Over a third of patients had normal LV function suggesting that cardiomyopathy is not inevitable in this group.
Collapse
Affiliation(s)
- Lauren O'Brien
- 1Department of Respiratory Medicine, West Park Healthcare Centre, Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
174
|
Mani H, Tanaka H, Adachi T, Ikegawa M, Dai P, Fujita N, Takamatsu T. How Does the Ca(2+)-paradox Injury Induce Contracture in the Heart?-A Combined Study of the Intracellular Ca(2+) Dynamics and Cell Structures in Perfused Rat Hearts. Acta Histochem Cytochem 2015; 48:1-8. [PMID: 25861132 PMCID: PMC4387258 DOI: 10.1267/ahc.14059] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 12/09/2014] [Indexed: 01/01/2023] Open
Abstract
The calcium (Ca2+)-paradox injury of the heart, induced by restoration of extracellular Ca2+ after its short-term depletion, is known to provoke cardiomyocyte contracture. However, undetermined is how the Ca2+-paradox provokes such a distinctive presentation of myocytes in the heart. To address this, we imaged sequential intracellular Ca2+ dynamics and concomitant structures of the subepicardial ventricular myocytes in fluo3-loaded, Langendorff-perfused rat hearts produced by the Ca2+ paradox. Under rapid-scanning confocal microscopy, repletion of Ca2+ following its depletion produced high-frequency Ca2+ waves in individual myocytes with asynchronous localized contractions, resulting in contracture within 10 min. Such alterations of myocytes were attenuated by 5-mM NiCl2, but not by verapamil, SEA0400, or combination of ryanodine and thapsigargin, indicating a contribution of non-specific transmembrane Ca2+ influx in the injury. However, saponin-induced membrane permeabilization of Ca2+ showed no apparent contracture despite the emergence of high-frequency Ca2+ waves, indicating an essential role of myocyte-myocyte and myocyte-extracellular matrix (ECM) mechanical connections in the Ca2+ paradox. In immunohistochemistry Ca2+ depletion produced separation of the intercalated disc that expresses cadherin and dissipation of β-dystroglycan located along the sarcolemma. Taken together, along with the trans-sarcolemmal Ca2+ influx, disruption of cell-cell and cell-ECM connections is essential for contracture in the Ca2+-paradox injury.
Collapse
Affiliation(s)
- Hiroki Mani
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Graduate School of Medical Science
- Department of Molecular Genetics and Laboratory Medicine, Kyoto Prefectural University of Medicine, Graduate School of Medical Science
| | - Hideo Tanaka
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Graduate School of Medical Science
| | - Tetsuya Adachi
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Graduate School of Medical Science
| | - Masaya Ikegawa
- Department of Genomic Medical Sciences, Kyoto Prefectural University of Medicine, Graduate School of Medical Science
| | - Ping Dai
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Graduate School of Medical Science
| | - Naohisa Fujita
- Department of Molecular Genetics and Laboratory Medicine, Kyoto Prefectural University of Medicine, Graduate School of Medical Science
| | - Tetsuro Takamatsu
- Department of Pathology and Cell Regulation, Kyoto Prefectural University of Medicine, Graduate School of Medical Science
| |
Collapse
|
175
|
Sharma P, Jha A, Stelmack GL, Detillieux K, Basu S, Klonisch T, Unruh H, Halayko AJ. Characterization of the dystrophin-glycoprotein complex in airway smooth muscle: role of δ-sarcoglycan in airway responsiveness. Can J Physiol Pharmacol 2015; 93:195-202. [PMID: 25692961 DOI: 10.1139/cjpp-2014-0389] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The dystrophin-glycoprotein complex (DGC) is an integral part of caveolae microdomains, and its interaction with caveolin-1 is essential for the phenotype and functional properties of airway smooth muscle (ASM). The sarcoglycan complex provides stability to the dystroglycan complex, but its role in ASM contraction and lung physiology in not understood. We tested whether δ-sarcoglycan (δ-SG), through its interaction with the DGC, is a determinant of ASM contraction ex vivo and airway mechanics in vivo. We measured methacholine (MCh)-induced isometric contraction and airway mechanics in δ-SG KO and wild-type mice. Last, we performed immunoblotting and transmission electron microscopy to assess DGC protein expression and the ultrastructural features of tracheal smooth muscle. Our results reveal an age-dependent reduction in the MCh-induced tracheal isometric force and significant reduction in airway resistance at high concentrations of MCh (50.0 mg/mL) in δ-SG KO mice. The changes in contraction and lung function correlated with decreased caveolin-1 and β-dystroglycan abundance, as well as an age-dependent loss of caveolae in the cell membrane of tracheal smooth muscle in δ-SG KO mice. Collectively, these results confirm and extend understanding of a functional role for the DGC in the contractile properties of ASM and demonstrate that this results in altered lung function in vivo.
Collapse
Affiliation(s)
- Pawan Sharma
- Departments of Physiology and Pathophysiology, University of Manitoba, John Buhler Research Centre, 715 McDermot Avenue, Winnipeg, MB R3E 3P4, Canada., Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | | | | | | | | | | | | | | |
Collapse
|
176
|
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
|
177
|
Rice KM, Manne NDPK, Gadde MK, Paturi S, Arvapalli R, Blough E. Differential regulation of apoptosis in slow and fast twitch muscles of aged female F344BN rats. AGE (DORDRECHT, NETHERLANDS) 2015; 37:30. [PMID: 25813803 PMCID: PMC4375133 DOI: 10.1007/s11357-015-9767-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 03/09/2015] [Indexed: 05/13/2023]
Abstract
Age-related muscle atrophy is characterized by decreases in muscle mass and is thought be mediated, at least in part, by increases in myocyte apoptosis. Recent data has demonstrated that the degree of muscle loss with aging may differ between males and females while other work has suggested that apoptosis as indicated by DNA fragmentation may be regulated differently in fast- and slow-twitch muscles. Herein, we investigate how aging affects the regulation of muscle apoptosis in the fast-twitch extensor digitorum longus (EDL) and slow-twitch soleus muscles of young (6-month), aged (26-month), and very aged (30-month) female Fischer 344/NNiaHSD × Brown Norway/BiNia (F344BN) rats. Tissue sections were stained with hydroethidium for ROS and protein extract was subjected to immunoblotting for assessing apoptotic markers. Our data suggest that decreases in muscle mass were associated with increased DNA fragmentation (TUNEL positive) and increases in reactive oxygen species (ROS) as determined by hydroethidium staining in both the EDL and soleus. Similar to our previous work using aged male animals, we observed that the time course and magnitude of changes in Bax, Bcl-2, caspase-3, caspase-9, and cleavage of α-fodrin protein were regulated differently between muscles. These data suggest that aging in the female F344BN rat is associated with decreases in muscle mass, elevations in ROS level, increased muscle cell DNA fragmentation, and alterations in cell membrane integrity and that apoptotic mechanisms may differ between fiber types.
Collapse
Affiliation(s)
- Kevin M Rice
- Center for Diagnostic Nanosystems, Marshall University, Room 241D, Robert C. Byrd Biotechnology Science Center Building, 1700 3rd Ave., Huntington, WV, 25755-1090, USA,
| | | | | | | | | | | |
Collapse
|
178
|
Rodríguez-Cruz M, Sanchez R, Escobar RE, Cruz-Guzmán ODR, López-Alarcón M, Bernabe García M, Coral-Vázquez R, Matute G, Velázquez Wong AC. Evidence of Insulin Resistance and Other Metabolic Alterations in Boys with Duchenne or Becker Muscular Dystrophy. Int J Endocrinol 2015; 2015:867273. [PMID: 26089900 PMCID: PMC4452344 DOI: 10.1155/2015/867273] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 08/29/2014] [Accepted: 08/30/2014] [Indexed: 01/29/2023] Open
Abstract
Aim. Our aim was (1) to determine the frequency of insulin resistance (IR) in patients with Duchenne/Becker muscular dystrophy (DMD/BMD), (2) to identify deleted exons of DMD gene associated with obesity and IR, and (3) to explore some likely molecular mechanisms leading to IR. Materials and Methods. In 66 patients with DMD/BMD without corticosteroids treatment, IR, obesity, and body fat mass were evaluated. Molecules involved in glucose metabolism were analyzed in muscle biopsies. Results show that 18.3%, 22.7%, and 68% were underweight, overweight, or obese, and with high adiposity, respectively; 48.5% and 36.4% presented hyperinsulinemia and IR, respectively. Underweight patients (27.3%) exhibited hyperinsulinemia and IR. Carriers of deletions in exons 45 (OR = 9.32; 95% CI = 1.16-74.69) and 50 (OR = 8.73; 95% CI = 1.17-65.10) from DMD gene presented higher risk for IR than noncarriers. We observed a greater staining of cytoplasmic aggregates for GLUT4 in muscle biopsies than healthy muscle tissue. Conclusion. Obesity, hyperinsulinemia, and IR were observed in DMD/BMD patients and are independent of corticosteroids treatment. Carriers of deletion in exons 45 or 50 from DMD gene are at risk for developing IR. It is suggested that alteration in GLUT4 in muscle fibers from DMD patients could be involved in IR.
Collapse
Affiliation(s)
- Maricela Rodríguez-Cruz
- Laboratorio de Biología Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, Apartado Postal C-029 C. S.P.I. “Coahuila”, Coahuila No. 5, Colonia Roma, 06703 México, DF, Mexico
- *Maricela Rodríguez-Cruz:
| | - Raúl Sanchez
- Laboratorio de Biología Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, Apartado Postal C-029 C. S.P.I. “Coahuila”, Coahuila No. 5, Colonia Roma, 06703 México, DF, Mexico
| | - Rosa E. Escobar
- Servicio de Electrodiagnóstico y Distrofia Muscular, Instituto Nacional de la Rehabilitación, México, DF, Mexico
| | - Oriana del Rocío Cruz-Guzmán
- Laboratorio de Biología Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, Apartado Postal C-029 C. S.P.I. “Coahuila”, Coahuila No. 5, Colonia Roma, 06703 México, DF, Mexico
| | - Mardia López-Alarcón
- Laboratorio de Biología Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, Apartado Postal C-029 C. S.P.I. “Coahuila”, Coahuila No. 5, Colonia Roma, 06703 México, DF, Mexico
| | - Mariela Bernabe García
- Laboratorio de Biología Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, Apartado Postal C-029 C. S.P.I. “Coahuila”, Coahuila No. 5, Colonia Roma, 06703 México, DF, Mexico
| | - Ramón Coral-Vázquez
- Sección de Estudios de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Plan de San Luis y Díaz Mirón s/n, Col. Casco de Santo Tomas, Delegación Miguel Hidalgo, 11340 México City, Mexico
- Subdirección de Enseñanza e Investigación, Centro Médico Nacional 20 de Noviembre, Instituto de Seguridad y Servicios Sociales de los Trabajadores del Estado, San Lorenzo 502 (2 Piso), Colonia Del Valle, Delegación Benito Juárez, 03100 México City, Mexico
| | - Guadalupe Matute
- Laboratorio de Biología Molecular, Unidad de Investigación Médica en Nutrición, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, Apartado Postal C-029 C. S.P.I. “Coahuila”, Coahuila No. 5, Colonia Roma, 06703 México, DF, Mexico
| | - Ana Claudia Velázquez Wong
- Unidad de Investigación Médica en Genética Humana, Hospital de Pediatría, Centro Médico Nacional Siglo XXI, IMSS, México, DF, Mexico
| |
Collapse
|
179
|
Brehm MA, Kempen JCE, van der Kooi AJ, de Groot IJM, van den Bergen JC, Verschuuren JJGM, Niks EH, Harlaar J. Age-related longitudinal changes in metabolic energy expenditure during walking in boys with Duchenne muscular dystrophy. PLoS One 2014; 9:e115200. [PMID: 25506914 PMCID: PMC4266650 DOI: 10.1371/journal.pone.0115200] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Accepted: 11/19/2014] [Indexed: 12/03/2022] Open
Abstract
Objective The aim of this study was to evaluate age-related changes in metabolic walking energy expenditure in ambulant boys affected by Duchenne muscular dystrophy over a follow-up period of 12 months. Methods At baseline (T1) and 12 months later (T2), metabolic walking energy expenditure was assessed during a 6-minute walk test at comfortable speed in 14 ambulant boys with Duchenne (age range: 6.0-12.5 years, mean 8.2). Outcome measures derived from the assessment included the 6-minute comfortable walking distance (m) and net-nondimensional energy cost relative to speed-matched control cost (SMC-EC, %). Statistical comparisons were made using a two-way repeated measures ANOVA (factors: time (T1 versus T2) and age (<8 years of age (yoa) versus ≥8 yoa)). Results Over the course of the study, a significant decrease of -28m (−8.2%, p = 0.043) was noted in the walked distance at comfortable speed. Besides, SMC-EC increased with 4.4%, although this change was not significant (p = 0.452). Regarding age groups, boys below 8 yoa showed a smaller annual decrease in the walked distance (−15 m) compared to boys above 8 yoa (−37 m). SMC-EC increased with 10% in the older boys, while in the younger boys it decreased (−2.1%). The main effect of age group on walking distance and SMC-EC however was not significant (p>0.158), and also there were no interaction effects (p>0.248). Conclusions The results of our small study suggest that the natural course of walking performance in ambulant boys with Duchenne is characterized by a decrease in comfortable walking distance and an increase in walking energy cost. The rate of energy cost seems to increase with age, while walking distance decreases, which is opposite from the trend in typically developing children.
Collapse
Affiliation(s)
- Merel-Anne Brehm
- Department of Rehabilitation Medicine and MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands
- Department of Rehabilitation, Academic Medical Center, University of Amsterdam, The Netherlands
- * E-mail:
| | - Jiska C. E. Kempen
- Department of Rehabilitation Medicine and MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands
| | - Anneke J. van der Kooi
- Department of Neurology, Academic Medical Center, University of Amsterdam, The Netherlands
| | - Imelda J. M. de Groot
- Department of Rehabilitation, Radboud University Medical Center Nijmegen, The Netherlands
| | | | | | - Erik H. Niks
- Department of Neurology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jaap Harlaar
- Department of Rehabilitation Medicine and MOVE Research Institute, VU University Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
180
|
Predictive value of myocardial delayed enhancement in Duchenne muscular dystrophy. Pediatr Cardiol 2014; 35:1279-85. [PMID: 24830760 DOI: 10.1007/s00246-014-0929-z] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2014] [Accepted: 04/29/2014] [Indexed: 01/28/2023]
Abstract
In other cardiomyopathies, cardiac magnetic resonance imaging (CMR)-derived myocardial delayed enhancement (MDE), a marker of myocardial fibrosis, is a risk factor for sudden cardiac death (SCD). In Duchenne muscular dystrophy (DMD), the prognostic value of MDE for ventricular arrhythmias and death is unknown. This study aimed to evaluate associations between MDE and electrocardiographic (ECG) changes, ventricular remodeling, risk of arrhythmias, and death in DMD. This retrospective study included all subjects with DMD who had undergone a CMR between January 2006 and December 2011 and had available ECG and 24-h Holter records from the same period. Left ventricular (LV) MDE was semiquantitatively graded from 0 to 4. Comparisons of demographic and clinical characteristics between MDE and no-MDE groups were made. Cox regression analysis was performed to assess factors associated with death. This study investigated 32 boys with a median age of 13.8 years (range, 7.2-17.4 years) and found MDE present in 25 (78 %) of the boys. Compared with the no-MDE subjects, the MDE subjects were older (15.7 ± 3.3 vs 12.1 ± 4.8 years) and had a wider QT dispersion (QTd: 74 ± 30 vs 55 ± 33 ms), a higher incidence of ventricular tachycardia (40 vs 0 %), a lower LV ejection fraction (46 ± 12 vs 56 ± 9 %), a larger LV end-diastolic volume (124 ± 58 vs 68 ± 14 ml/m(2)), and a larger end-systolic volume (57 ± 29 vs 28 ± 10 ml/m(2)) (p < 0.05 for all). During the study period, six of the subjects (19 %) died. The factors associated with mortality were increased age, advanced grade of MDE, higher LV end-systolic volume, lower LV ejection fraction, use of beta-blockers, and ventricular tachycardia. Myocardial fibrosis detected by CMR is an independent predictor of adverse cardiac remodeling, ventricular arrhythmias, and death in DMD. Cardiac MRI using MDE can be applied as a screening tool to detect patients at risk for ventricular arrhythmias, more advanced disease, adverse LV remodeling, and death.
Collapse
|
181
|
Sharma P, Basu S, Mitchell RW, Stelmack GL, Anderson JE, Halayko AJ. Role of dystrophin in airway smooth muscle phenotype, contraction and lung function. PLoS One 2014; 9:e102737. [PMID: 25054970 PMCID: PMC4108318 DOI: 10.1371/journal.pone.0102737] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 06/23/2014] [Indexed: 11/19/2022] Open
Abstract
Dystrophin links the transmembrane dystrophin-glycoprotein complex to the actin cytoskeleton. We have shown that dystrophin-glycoprotein complex subunits are markers for airway smooth muscle phenotype maturation and together with caveolin-1, play an important role in calcium homeostasis. We tested if dystrophin affects phenotype maturation, tracheal contraction and lung physiology. We used dystrophin deficient Golden Retriever dogs (GRMD) and mdx mice vs healthy control animals in our approach. We found significant reduction of contractile protein markers: smooth muscle myosin heavy chain (smMHC) and calponin and reduced Ca2+ response to contractile agonist in dystrophin deficient cells. Immunocytochemistry revealed reduced stress fibers and number of smMHC positive cells in dystrophin-deficient cells, when compared to control. Immunoblot analysis of Akt1, GSK3β and mTOR phosphorylation further revealed that downstream PI3K signaling, which is essential for phenotype maturation, was suppressed in dystrophin deficient cell cultures. Tracheal rings from mdx mice showed significant reduction in the isometric contraction to methacholine (MCh) when compared to genetic control BL10ScSnJ mice (wild-type). In vivo lung function studies using a small animal ventilator revealed a significant reduction in peak airway resistance induced by maximum concentrations of inhaled MCh in mdx mice, while there was no change in other lung function parameters. These data show that the lack of dystrophin is associated with a concomitant suppression of ASM cell phenotype maturation in vitro, ASM contraction ex vivo and lung function in vivo, indicating that a linkage between the DGC and the actin cytoskeleton via dystrophin is a determinant of the phenotype and functional properties of ASM.
Collapse
MESH Headings
- Animals
- Blotting, Western
- Cells, Cultured
- Dogs
- Dystrophin/deficiency
- Dystrophin/genetics
- Dystrophin/physiology
- Immunohistochemistry
- Lung/metabolism
- Lung/physiology
- Methacholine Chloride/pharmacology
- Mice, Inbred mdx
- Mice, Knockout
- Microscopy, Electron, Transmission
- Microscopy, Fluorescence
- Muscle Contraction/genetics
- Muscle Contraction/physiology
- Muscle, Smooth/cytology
- Muscle, Smooth/metabolism
- Muscle, Smooth/physiology
- Myocytes, Smooth Muscle/cytology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/physiology
- Myosin Heavy Chains/metabolism
- Phosphatidylinositol 3-Kinases/metabolism
- Respiratory System/cytology
- Respiratory System/metabolism
- Respiratory System/ultrastructure
- Signal Transduction/genetics
- Signal Transduction/physiology
- Trachea/drug effects
- Trachea/metabolism
- Trachea/physiology
Collapse
Affiliation(s)
- Pawan Sharma
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
- CIHR National Training Program in Allergy and Asthma, University of Manitoba, Winnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Sujata Basu
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Richard W. Mitchell
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Gerald L. Stelmack
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
| | - Judy E. Anderson
- Department of Biological Sciences, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Andrew J. Halayko
- Department of Physiology and Pathophysiology, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
- Section of Respiratory Disease, University of Manitoba, Winnipeg, Manitoba, Canada
- CIHR National Training Program in Allergy and Asthma, University of Manitoba, Winnipeg, Manitoba, Canada
- Biology of Breathing Group, Manitoba Institute of Child Health, Winnipeg, Manitoba, Canada
- * E-mail:
| |
Collapse
|
182
|
From innate to adaptive immune response in muscular dystrophies and skeletal muscle regeneration: the role of lymphocytes. BIOMED RESEARCH INTERNATIONAL 2014; 2014:438675. [PMID: 25028653 PMCID: PMC4083765 DOI: 10.1155/2014/438675] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 05/02/2014] [Indexed: 12/04/2022]
Abstract
Skeletal muscle is able to restore contractile functionality after injury thanks to its ability to regenerate. Following muscle necrosis, debris is removed by macrophages, and muscle satellite cells (MuSCs), the muscle stem cells, are activated and subsequently proliferate, migrate, and form muscle fibers restoring muscle functionality. In most muscle dystrophies (MDs), MuSCs fail to properly proliferate, differentiate, or replenish the stem cell compartment, leading to fibrotic deposition. However, besides MuSCs, interstitial nonmyogenic cells and inflammatory cells also play a key role in orchestrating muscle repair. A complete understanding of the complexity of these mechanisms should allow the design of interventions to attenuate MDs pathology without disrupting regenerative processes. In this review we will focus on the contribution of immune cells in the onset and progression of MDs, with particular emphasis on Duchenne muscular dystrophy (DMD). We will briefly summarize the current knowledge and recent advances made in our understanding of the involvement of different innate immune cells in MDs and will move on to critically evaluate the possible role of cell populations within the acquired immune response. Revisiting previous observations in the light of recent evidence will likely change our current view of the onset and progression of the disease.
Collapse
|
183
|
Vaughan EM, You JS, Elsie Yu HY, Lasek A, Vitale N, Hornberger TA, Bement WM. Lipid domain-dependent regulation of single-cell wound repair. Mol Biol Cell 2014; 25:1867-76. [PMID: 24790096 PMCID: PMC4055266 DOI: 10.1091/mbc.e14-03-0839] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 04/14/2014] [Accepted: 04/21/2014] [Indexed: 11/21/2022] Open
Abstract
After damage, cells reseal their plasma membrane and repair the underlying cortical cytoskeleton. Although many different proteins have been implicated in cell repair, the potential role of specific lipids has not been explored. Here we report that cell damage elicits rapid formation of spatially organized lipid domains around the damage site, with different lipids concentrated in different domains as a result of both de novo synthesis and transport. One of these lipids-diacylglycerol (DAG)-rapidly accumulates in a broad domain that overlaps the zones of active Rho and Cdc42, GTPases that regulate repair of the cortical cytoskeleton. Formation of the DAG domain is required for Cdc42 and Rho activation and healing. Two DAG targets, protein kinase C (PKC) β and η, are recruited to cell wounds and play mutually antagonistic roles in the healing process: PKCβ participates in Rho and Cdc42 activation, whereas PKCη inhibits Rho and Cdc42 activation. The results reveal an unexpected diversity in subcellular lipid domains and the importance of such domains for a basic cellular process.
Collapse
Affiliation(s)
- Emily M Vaughan
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706Department of Zoology, University of Wisconsin-Madison, Madison, WI 53706
| | - Jae-Sung You
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706
| | - Hoi-Ying Elsie Yu
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706Department of Zoology, University of Wisconsin-Madison, Madison, WI 53706
| | - Amber Lasek
- Department of Zoology, University of Wisconsin-Madison, Madison, WI 53706
| | - Nicolas Vitale
- Institut des Neurosciences Cellulaires et Integratives, Centre National de la Recherche Scientifique UPR 3212, and Université de Strasbourg, 67400 Strasbourg, France
| | - Troy A Hornberger
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706Department of Comparative Biosciences, University of Wisconsin-Madison, Madison, WI 53706
| | - William M Bement
- Program in Cellular and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706Department of Zoology, University of Wisconsin-Madison, Madison, WI 53706Laboratory of Cell and Molecular Biology, University of Wisconsin-Madison, Madison, WI 53706
| |
Collapse
|
184
|
Wissing ER, Boyer JG, Kwong JQ, Sargent MA, Karch J, McNally EM, Otsu K, Molkentin JD. P38α MAPK underlies muscular dystrophy and myofiber death through a Bax-dependent mechanism. Hum Mol Genet 2014; 23:5452-63. [PMID: 24876160 DOI: 10.1093/hmg/ddu270] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Muscular dystrophies are a group of genetic diseases that lead to muscle wasting and, in most cases, premature death. Cytokines and inflammatory factors are released during the disease process where they promote deleterious signaling events that directly participate in myofiber death. Here, we show that p38α, a kinase in the greater mitogen-activated protein kinase (MAPK)-signaling network, serves as a nodal regulator of disease signaling in dystrophic muscle. Deletion of Mapk14 (p38α-encoding gene) in the skeletal muscle of mdx- (lacking dystrophin) or sgcd- (δ-sarcoglycan-encoding gene) null mice resulted in a significant reduction in pathology up to 6 months of age. We also generated MAPK kinase 6 (MKK6) muscle-specific transgenic mice to model heightened p38α disease signaling that occurs in dystrophic muscle, which resulted in severe myofiber necrosis and many hallmarks of muscular dystrophy. Mechanistically, we show that p38α directly induces myofiber death through a mitochondrial-dependent pathway involving direct phosphorylation and activation of the pro-death Bcl-2 family member Bax. Indeed, muscle-specific deletion of Bax, but not the apoptosis regulatory gene Tp53 (encoding p53), significantly reduced dystrophic pathology in the muscles of MKK6 transgenic mice. Moreover, use of a p38 MAPK pharmacologic inhibitor reduced dystrophic disease in Sgcd(-/-) mice suggesting a future therapeutic approach to delay disease.
Collapse
Affiliation(s)
- Erin R Wissing
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| | - Justin G Boyer
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| | - Jennifer Q Kwong
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| | - Michelle A Sargent
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| | - Jason Karch
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| | - Elizabeth M McNally
- Department of Medicine, Section of Cardiology, University of Chicago, 5841 S. Maryland, MC 6088, Chicago, IL 60637, USA
| | - Kinya Otsu
- Cardiovascular Division, King's College London, 125 Coldharbour Lane, London SE5 9NU, UK and
| | - Jeffery D Molkentin
- Department of Pediatrics, Cincinnati Children's Hospital Medical Center, University of Cincinnati, Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA, Howard Hughes Medical Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, 240 Albert Sabin Way, Cincinnati, OH 45229, USA
| |
Collapse
|
185
|
Chronic oral administration of Ang-(1-7) improves skeletal muscle, autonomic and locomotor phenotypes in muscular dystrophy. Clin Sci (Lond) 2014; 127:101-9. [PMID: 24502705 DOI: 10.1042/cs20130602] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Muscular dystrophies are a group of heterogeneous genetic disorders that cause progressive muscle weakness and wasting, dilated cardiomyopathy and early mortality. There are different types of muscular dystrophies with varying aetiologies but they all have a common hallmark of myofibre degeneration, atrophy and decreased mobility. Mutation in Sgcd (sarcoglycan-δ), a subunit of dystrophin glycoprotein complex, causes LGMD2F (limb girdle muscular dystrophy 2F). Previously, we have reported that Sgcd-deficient (Sgcd-/-) mice exhibit AngII (angiotensin II)-induced autonomic and skeletal muscle dysfunction at a young age, which contributes to onset of dilated cardiomyopathy and mortality at older ages. Two counter-regulatory RAS (renin-angiotensin system) pathways have been identified: deleterious actions of AngII acting on the AT1R (AngII type 1 receptor) compared with the protective actions of Ang-(1-7) [angiotensin-(1-7)] acting on the receptor Mas. We propose that the balance between the AngII/AT1R and Ang-(1-7)/Mas axes is disturbed in Sgcd-/- mice. Control C57BL/6J and Sgcd-/- mice were treated with Ang-(1-7) included in hydroxypropyl β-cyclodextrin (in drinking water) for 8-9 weeks beginning at 3 weeks of age. Ang-(1-7) treatment restored the AngII/AT1R compared with Ang-(1-7)/Mas balance, decreased oxidative stress and fibrosis in skeletal muscle, increased locomotor activity, and prevented autonomic dysfunction without lowering blood pressure in Sgcd-/- mice. Our results suggest that correcting the early autonomic dysregulation by administering Ang-(1-7) or enhancing its endogenous production may provide a novel therapeutic approach in muscular dystrophy.
Collapse
|
186
|
McDade JR, Archambeau A, Michele DE. Rapid actin-cytoskeleton-dependent recruitment of plasma membrane-derived dysferlin at wounds is critical for muscle membrane repair. FASEB J 2014; 28:3660-70. [PMID: 24784578 DOI: 10.1096/fj.14-250191] [Citation(s) in RCA: 69] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Deficits in membrane repair may contribute to disease progression in dysferlin-deficient muscular dystrophy. Dysferlin, a type-II transmembrane phospholipid-binding protein, is hypothesized to regulate fusion of repair vesicles with the sarcolemma to facilitate membrane repair, but the dysferlin-containing compartments involved in membrane repair and the mechanism by which these compartments contribute to resealing are unclear. A dysferlin-pHluorin [dysf-pH-sensitive green fluorescent protein (pHGFP)] muscle-specific transgenic mouse was developed to examine the dynamic behavior and subcellular localization of dysferlin during membrane repair in adult skeletal muscle fibers. Live-cell confocal microscopy of uninjured adult dysf-pHGFP muscle fibers revealed that dysferlin is highly enriched in the sarcolemma and transverse tubules. Laser-wounding induced rapid recruitment of ∼30 μm of local dysferlin-containing sarcolemma, leading to formation of stable dysferlin accumulations surrounding lesions, endocytosis of dysferlin, and formation of large cytoplasmic vesicles from distal regions of the fiber. Disruption of the actin cytoskeleton decreased recruitment of sarcolemma-derived dysferlin to lesions in dysf-pHGFP fibers without affecting endocytosis and impaired membrane resealing in wild-type fibers, similar to findings in dysferlin deficiency (a 2-fold increase in FM1-43 uptake). Our data support a new mechanism whereby recruitment of sarcolemma-derived dysferlin creates an active zone of high lipid-binding activity at wounds to interact with repair vesicles and facilitate membrane resealing in skeletal muscle.
Collapse
Affiliation(s)
- Joel R McDade
- Department of Molecular and Integrative Physiology and
| | | | - Daniel E Michele
- Department of Molecular and Integrative Physiology and Department of Internal Medicine, Division of Molecular Medicine and Genetics, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
187
|
Lu A, Poddar M, Tang Y, Proto JD, Sohn J, Mu X, Oyster N, Wang B, Huard J. Rapid depletion of muscle progenitor cells in dystrophic mdx/utrophin-/- mice. Hum Mol Genet 2014; 23:4786-800. [PMID: 24781208 DOI: 10.1093/hmg/ddu194] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) patients lack dystrophin from birth; however, muscle weakness becomes apparent only at 3-5 years of age, which happens to coincide with the depletion of the muscle progenitor cell (MPC) pools. Indeed, MPCs isolated from older DMD patients demonstrate impairments in myogenic potential. To determine whether the progression of muscular dystrophy is a consequence of the decline in functional MPCs, we investigated two animal models of DMD: (i) dystrophin-deficient mdx mice, the most commonly utilized model of DMD, which has a relatively mild dystrophic phenotype and (ii) dystrophin/utrophin double knock-out (dKO) mice, which display a similar histopathologic phenotype to DMD patients. In contrast to age-matched mdx mice, we observed that both the number and regeneration potential of dKO MPCs rapidly declines during disease progression. This occurred in MPCs at both early and late stages of myogenic commitment. In fact, early MPCs isolated from 6-week-old dKO mice have reductions in proliferation, resistance to oxidative stress and multilineage differentiation capacities compared with age-matched mdx MPCs. This effect may potentially be mediated by fibroblast growth factor overexpression and/or a reduction in telomerase activity. Our results demonstrate that the rapid disease progression in the dKO model is associated, at least in part, with MPC depletion. Therefore, alleviating MPC depletion could represent an approach to delay the onset of the histopathologies associated with DMD patients.
Collapse
Affiliation(s)
- Aiping Lu
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Minakshi Poddar
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Ying Tang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Jonathan D Proto
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Jihee Sohn
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Xiaodong Mu
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Nicholas Oyster
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Bing Wang
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Johnny Huard
- Department of Orthopaedic Surgery, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
188
|
Abstract
Many monogenic disorders, including the muscular dystrophies, display phenotypic variability despite the same disease-causing mutation. To identify genetic modifiers of muscular dystrophy and its associated cardiomyopathy, we used quantitative trait locus mapping and whole genome sequencing in a mouse model. This approach uncovered a modifier locus on chromosome 11 associated with sarcolemmal membrane damage and heart mass. Whole genome and RNA sequencing identified Anxa6, encoding annexin A6, as a modifier gene. A synonymous variant in exon 11 creates a cryptic splice donor, resulting in a truncated annexin A6 protein called ANXA6N32. Live cell imaging showed that annexin A6 orchestrates a repair zone and cap at the site of membrane disruption. In contrast, ANXA6N32 dramatically disrupted the annexin A6-rich cap and the associated repair zone, permitting membrane leak. Anxa6 is a modifier of muscular dystrophy and membrane repair after injury.
Collapse
|
189
|
Altered acetylcholine release in the hippocampus of dystrophin-deficient mice. Neuroscience 2014; 269:173-83. [PMID: 24704431 DOI: 10.1016/j.neuroscience.2014.03.050] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2014] [Revised: 03/22/2014] [Accepted: 03/25/2014] [Indexed: 12/29/2022]
Abstract
Mild cognitive impairments have been described in one-third of patients with Duchenne muscle dystrophy (DMD). DMD is characterized by progressive and irreversible muscle degeneration caused by mutations in the dystrophin gene and lack of the protein expression. Previously, we have reported altered concentrations of α7- and β2-containing nicotinic acetylcholine receptors (nAChRs) in hippocampal membranes of dystrophic (mdx) mice. This suggests that alterations in the central cholinergic synapses are associated with dystrophin deficiency. In this study, we examined the release of acetylcholine (ACh) and the level of the vesicular ACh transporter (VAChT) using synaptosomes isolated from brain regions that normally have a high density of dystrophin (cortex, hippocampus and cerebellum), in control and mdx mice at 4 and 12months of age. ACh release evoked by nicotinic stimulation or K(+) depolarization was measured as the tritium outflow from superfused synaptosomes preloaded with [(3)H]-choline. The results showed that the evoked tritium release was Ca(2+)-dependent and mostly formed by [(3)H]-ACh. β2-containing nAChRs were involved in agonist-evoked [(3)H]-ACh release in control and mdx preparations. In hippocampal synaptosomes from 12-month-old mdx mice, nAChR-evoked [(3)H]-ACh release increased by 57% compared to age-matched controls. Moreover, there was a 98% increase in [(3)H]-ACh release compared to 4-month-old mdx mice. [(3)H]-ACh release evoked by K(+) depolarization was not altered, while the VAChT protein level was decreased (19%) compared to that of age-matched controls. In cortical and cerebellar preparations, there was no difference in nAChR-evoked [(3)H]-ACh release and VAChT levels between mdx and age-matched control groups. Our previous findings and the presynaptic alterations observed in the hippocampi of 12-month-old mdx mice indicate possible dysfunction of nicotinic cholinergic synapses associated with dystrophin deficiency. These changes may contribute to the cognitive and behavioral abnormalities described in dystrophic mice and patients with DMD.
Collapse
|
190
|
Peikert K, Kasper M, May CA. Distribution of caveolin in the muscle spindles of human skeletal muscle. J Anat 2014; 224:681-7. [PMID: 24660982 DOI: 10.1111/joa.12173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2014] [Indexed: 01/16/2023] Open
Abstract
The aim of the present study was to demonstrate the location of the different members of the caveolin (cav) family in human muscle spindles. Twenty spindles of three human muscles (vastus medialis, ischiocavernosus, bulbospongiosus) from 12 cadavers were immunohistochemically stained for cav-1, cav-2, and cav-3, and the equatorial and polar regions evaluated. All layers of the outer and inner spindle capsule and all blood vessels within the spindle stained for cav-1 and cav-2. In the muscle spindle, intrafusal muscle fibres stained selectively for cav-3, but with a patchy appearance. Caveolinopathies may therefore also include changes in muscle spindle function.
Collapse
Affiliation(s)
- Kevin Peikert
- Department of Anatomy, Medical Faculty Carl Gustav Carus, TU Dresden, Dresden, Germany
| | | | | |
Collapse
|
191
|
Proteomic profiling of the dystrophin-deficient mdx phenocopy of dystrophinopathy-associated cardiomyopathy. BIOMED RESEARCH INTERNATIONAL 2014; 2014:246195. [PMID: 24772416 PMCID: PMC3977469 DOI: 10.1155/2014/246195] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/04/2013] [Accepted: 02/16/2014] [Indexed: 01/07/2023]
Abstract
Cardiorespiratory complications are frequent symptoms of Duchenne muscular dystrophy, a neuromuscular disorder caused by primary abnormalities in the dystrophin gene. Loss of cardiac dystrophin initially leads to changes in dystrophin-associated glycoproteins and subsequently triggers secondarily sarcolemmal disintegration, fibre necrosis, fibrosis, fatty tissue replacement, and interstitial inflammation. This results in progressive cardiac disease, which is the cause of death in a considerable number of patients afflicted with X-linked muscular dystrophy. In order to better define the molecular pathogenesis of this type of cardiomyopathy, several studies have applied mass spectrometry-based proteomics to determine proteome-wide alterations in dystrophinopathy-associated cardiomyopathy. Proteomic studies included both gel-based and label-free mass spectrometric surveys of dystrophin-deficient heart muscle from the established mdx animal model of dystrophinopathy. Comparative cardiac proteomics revealed novel changes in proteins associated with mitochondrial energy metabolism, glycolysis, signaling, iron binding, antibody response, fibre contraction, basal lamina stabilisation, and cytoskeletal organisation. This review summarizes the importance of studying cardiomyopathy within the field of muscular dystrophy research, outlines key features of the mdx heart and its suitability as a model system for studying cardiac pathogenesis, and discusses the impact of recent proteomic findings for exploring molecular and cellular aspects of cardiac abnormalities in inherited muscular dystrophies.
Collapse
|
192
|
Atypical phenotype in two patients with LAMA2 mutations. Neuromuscul Disord 2014; 24:419-24. [PMID: 24534542 DOI: 10.1016/j.nmd.2014.01.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Revised: 12/17/2013] [Accepted: 01/09/2014] [Indexed: 11/21/2022]
Abstract
Congenital muscular dystrophy type 1A is caused by mutations in the LAMA2 gene, which encodes the α2-chain of laminin. We report two patients with partial laminin-α2 deficiency and atypical phenotypes, one with almost exclusive central nervous system involvement (cognitive impairment and refractory epilepsy) and the second with marked cardiac dysfunction, rigid spine syndrome and limb-girdle weakness. Patients underwent clinical, histopathological, imaging and genetic studies. Both cases have two heterozygous LAMA2 variants sharing a potentially pathogenic missense mutation c.2461A>C (p.Thr821Pro) located in exon 18. Brain MRI was instrumental for the diagnosis, since muscular examination and motor achievements were normal in the first patient and there was a severe cardiac involvement in the second. The clinical phenotype of the patients is markedly different which could in part be explained by the different combination of mutations types (two missense versus a missense and a truncating mutation).
Collapse
|
193
|
Mezghenna K, Leroy J, Azay-Milhau J, Tousch D, Castex F, Gervais S, Delgado-Betancourt V, Gross R, Lajoix AD. Counteracting neuronal nitric oxide synthase proteasomal degradation improves glucose transport in insulin-resistant skeletal muscle from Zucker fa/fa rats. Diabetologia 2014; 57:177-86. [PMID: 24186360 DOI: 10.1007/s00125-013-3084-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 09/30/2013] [Indexed: 12/18/2022]
Abstract
AIMS/HYPOTHESIS Insulin-mediated glucose transport and utilisation are decreased in skeletal muscle from type 2 diabetic and glucose-intolerant individuals because of alterations in insulin receptor signalling, GLUT4 translocation to the plasma membrane and microvascular blood flow. Catalytic activity of the muscle-specific isoform of neuronal nitric oxide synthase (nNOS) also participates in the regulation of glucose transport and appears to be decreased in a relevant animal model of drastic insulin resistance, the obese Zucker fa/fa rat. Our objective was to determine the molecular mechanisms involved in this defect. METHODS Isolated rat muscles and primary cultures of myocytes were used for western blot analysis of protein expression, immunohistochemistry, glucose uptake measurements and GLUT4 translocation assays. RESULTS nNOS expression was reduced in skeletal muscle from fa/fa rats. This was caused by increased ubiquitination of the enzyme and subsequent degradation by the ubiquitin proteasome pathway. The degradation occurred through a greater interaction of nNOS with the chaperone heat-shock protein 70 and the co-chaperone, carboxyl terminus of Hsc70-interacting protein (CHIP). In addition, an alteration in nNOS sarcolemmal localisation was observed. We confirmed the implication of nNOS breakdown in defective insulin-induced glucose transport by demonstrating that blockade of proteasomal degradation or overexpression of nNOS improved basal and/or insulin-stimulated glucose uptake and GLUT4 translocation in primary cultures of insulin-resistant myocytes. CONCLUSIONS/INTERPRETATION Recovery of nNOS in insulin-resistant muscles should be considered a potential new approach to address insulin resistance.
Collapse
Affiliation(s)
- Karima Mezghenna
- Centre for Pharmacology and Innovation in Diabetes, University Montpellier 1, EA 7288, 15 Avenue Charles Flahault, BP 14491, 34093, Montpellier cedex 5, France
| | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Limongelli G, D’Alessandro R, Maddaloni V, Rea A, Sarkozy A, McKenna WJ. Skeletal muscle involvement in cardiomyopathies. J Cardiovasc Med (Hagerstown) 2013; 14:837-61. [DOI: 10.2459/jcm.0b013e3283641c69] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
195
|
Horstick EJ, Gibbs EM, Li X, Davidson AE, Dowling JJ. Analysis of embryonic and larval zebrafish skeletal myofibers from dissociated preparations. J Vis Exp 2013:e50259. [PMID: 24300240 DOI: 10.3791/50259] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
The zebrafish has proven to be a valuable model system for exploring skeletal muscle function and for studying human muscle diseases. Despite the many advantages offered by in vivo analysis of skeletal muscle in the zebrafish, visualizing the complex and finely structured protein milieu responsible for muscle function, especially in whole embryos, can be problematic. This hindrance stems from the small size of zebrafish skeletal muscle (60 μm) and the even smaller size of the sarcomere. Here we describe and demonstrate a simple and rapid method for isolating skeletal myofibers from zebrafish embryos and larvae. We also include protocols that illustrate post preparation techniques useful for analyzing muscle structure and function. Specifically, we detail the subsequent immunocytochemical localization of skeletal muscle proteins and the qualitative analysis of stimulated calcium release via live cell calcium imaging. Overall, this video article provides a straight-forward and efficient method for the isolation and characterization of zebrafish skeletal myofibers, a technique which provides a conduit for myriad subsequent studies of muscle structure and function.
Collapse
Affiliation(s)
- Eric J Horstick
- Departments of Pediatrics and Neurology, University of Michigan
| | | | | | | | | |
Collapse
|
196
|
Lim BK, Peter AK, Xiong D, Narezkina A, Yung A, Dalton ND, Hwang KK, Yajima T, Chen J, Knowlton KU. Inhibition of Coxsackievirus-associated dystrophin cleavage prevents cardiomyopathy. J Clin Invest 2013; 123:5146-51. [PMID: 24200690 DOI: 10.1172/jci66271] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Accepted: 09/05/2013] [Indexed: 01/10/2023] Open
Abstract
Heart failure in children and adults is often the consequence of myocarditis associated with Coxsackievirus (CV) infection. Upon CV infection, enteroviral protease 2A cleaves a small number of host proteins including dystrophin, which links actin filaments to the plasma membrane of muscle fiber cells (sarcolemma). It is unknown whether protease 2A-mediated cleavage of dystrophin and subsequent disruption of the sarcolemma play a role in CV-mediated myocarditis. We generated knockin mice harboring a mutation at the protease 2A cleavage site of the dystrophin gene, which prevents dystrophin cleavage following CV infection. Compared with wild-type mice, we found that mice expressing cleavage-resistant dystrophin had a decrease in sarcolemmal disruption and cardiac virus titer following CV infection. In addition, cleavage-resistant dystrophin inhibited the cardiomyopathy induced by cardiomyocyte-restricted expression of the CV protease 2A transgene. These findings indicate that protease 2A-mediated cleavage of dystrophin is critical for viral propagation, enteroviral-mediated cytopathic effects, and the development of cardiomyopathy.
Collapse
|
197
|
Janke A, Upadhaya R, Snow WM, Anderson JE. A new look at cytoskeletal NOS-1 and β-dystroglycan changes in developing muscle and brain in control and mdx dystrophic mice. Dev Dyn 2013; 242:1369-81. [PMID: 23940011 DOI: 10.1002/dvdy.24031] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2013] [Revised: 07/17/2013] [Accepted: 07/25/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Loss of dystrophin profoundly affects muscle function and cognition. Changes in the dystrophin-glycoprotein complex (DGC) including disruption of nitric oxide synthase (NOS-1) may result from loss of dystrophin or secondarily after muscle damage. Disruptions in NOS-1 and beta-dystroglycan (bDG) were examined in developing diaphragm, quadriceps, and two brain regions between control and mdx mice at embryonic day E18 and postnatal days P1, P10, and P28. Age-dependent differential muscle loading allowed us to test the hypothesis that DGC changes are dependent on muscle use. RESULTS Muscle development, including loss of central nucleation and the localization of NOS-1 and bDG, was earlier in diaphragm than quadriceps; these features were differentially disrupted in dystrophic muscles. The NOS-1/bDG ratio, an index of DGC stability, was higher in dystrophic diaphragm (P10-P28) and quadriceps (P28) than controls. There were also distinct regional differences in NOS-1 and bDG in brain tissues with age and strain. NOS-1 increased with age in control forebrain and cerebellum, and in mdx cerebellum; NOS-1 and bDG were higher in control than mdx mouse forebrain. CONCLUSIONS Important developmental changes in structure and muscle DGC preceded the hallmarks of dystrophy, and are consistent with the impact of muscle-specific differential loading during maturation.
Collapse
Affiliation(s)
- Alyssa Janke
- Faculty of Science, Department of Biological Sciences, Faculty of Medicine, University of Manitoba, Winnipeg, Canada
| | | | | | | |
Collapse
|
198
|
Moloughney JG, Weisleder N. Poloxamer 188 (p188) as a membrane resealing reagent in biomedical applications. Recent Pat Biotechnol 2013; 6:200-11. [PMID: 23092436 DOI: 10.2174/1872208311206030200] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2012] [Revised: 07/30/2012] [Accepted: 08/04/2012] [Indexed: 11/22/2022]
Abstract
Maintenance of the integrity of the plasma membrane is essential for maintenance of cellular function and prevention of cell death. Since the plasma membrane is frequently exposed to a variety of mechanical and chemical insults the cell has evolved active processes to defend against these injuries by resealing disruptions in the plasma membrane. Cell membrane repair is a conserved process observed in nearly every cell type where intracellular vesicles are recruited to sites of membrane disruption where they can fuse with themselves or the plasma membrane to create a repair patch. When disruptions are extensive or there is an underlying pathology that reduces the membrane repair capacity of a cell this defense mechanism may prove insufficient and the cell could die due to breakdown of the plasma membrane. Extensive loss of cells can compromise the integrity and function of tissues and leading to disease. Thus, methods to increase membrane resealing capacity could have broad utility in a number of disease states. Efforts to find reagents that can modulate plasma membrane reseal found that specific tri-block copolymers, such as poloxamer 188 (P188, or Pluronic F68), can increase the structural stability and resealing of the plasma membrane. Here we review several current patents and patent applications that present inventions making use of P188 and other copolymers to treat specific disease states such as muscular dystrophy, heart failure, neurodegenerative disorders and electrical injuries, or to facilitate biomedical applications such as transplantation. There appears to be promise for the application of poloxamers in the treatment of various diseases, however there are potential concerns with toxicity with long term application and bioavailability in some cases.
Collapse
Affiliation(s)
- Joseph G Moloughney
- Department of Neuroscience and Cell Biology, UMDNJ- Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | |
Collapse
|
199
|
Nguyen-Tran DH, Hait NC, Sperber H, Qi J, Fischer K, Ieronimakis N, Pantoja M, Hays A, Allegood J, Reyes M, Spiegel S, Ruohola-Baker H. Molecular mechanism of sphingosine-1-phosphate action in Duchenne muscular dystrophy. Dis Model Mech 2013; 7:41-54. [PMID: 24077965 PMCID: PMC3882047 DOI: 10.1242/dmm.013631] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Duchenne muscular dystrophy (DMD) is a lethal muscle-wasting disease. Studies in Drosophila showed that genetic increase of the levels of the bioactive sphingolipid sphingosine-1-phosphate (S1P) or delivery of 2-acetyl-5-tetrahydroxybutyl imidazole (THI), an S1P lyase inhibitor, suppresses dystrophic muscle degeneration. In the dystrophic mouse (mdx), upregulation of S1P by THI increases regeneration and muscle force. S1P can act as a ligand for S1P receptors and as a histone deacetylase (HDAC) inhibitor. Because Drosophila has no identified S1P receptors and DMD correlates with increased HDAC2 levels, we tested whether S1P action in muscle involves HDAC inhibition. Here we show that beneficial effects of THI treatment in mdx mice correlate with significantly increased nuclear S1P, decreased HDAC activity and increased acetylation of specific histone residues. Importantly, the HDAC2 target microRNA genes miR-29 and miR-1 are significantly upregulated, correlating with the downregulation of the miR-29 target Col1a1 in the diaphragm of THI-treated mdx mice. Further gene expression analysis revealed a significant THI-dependent decrease in inflammatory genes and increase in metabolic genes. Accordingly, S1P levels and functional mitochondrial activity are increased after THI treatment of differentiating C2C12 cells. S1P increases the capacity of the muscle cell to use fatty acids as an energy source, suggesting that THI treatment could be beneficial for the maintenance of energy metabolism in mdx muscles.
Collapse
Affiliation(s)
- Diem-Hang Nguyen-Tran
- Molecular and Cellular Biology Graduate Program, University of Washington, Seattle, WA 98195-7350, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
200
|
D'Arcy CE, Feeney SJ, McLean CA, Gehrig SM, Lynch GS, Smith JE, Cowling BS, Mitchell CA, McGrath MJ. Identification of FHL1 as a therapeutic target for Duchenne muscular dystrophy. Hum Mol Genet 2013; 23:618-36. [PMID: 24087791 DOI: 10.1093/hmg/ddt449] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Utrophin is a potential therapeutic target for the fatal muscle disease, Duchenne muscular dystrophy (DMD). In adult skeletal muscle, utrophin is restricted to the neuromuscular and myotendinous junctions and can compensate for dystrophin loss in mdx mice, a mouse model of DMD, but requires sarcolemmal localization. NFATc1-mediated transcription regulates utrophin expression and the LIM protein, FHL1 which promotes muscle hypertrophy, is a transcriptional activator of NFATc1. By generating mdx/FHL1-transgenic mice, we demonstrate that FHL1 potentiates NFATc1 activation of utrophin to ameliorate the dystrophic pathology. Transgenic FHL1 expression increased sarcolemmal membrane stability, reduced muscle degeneration, decreased inflammation and conferred protection from contraction-induced injury in mdx mice. Significantly, FHL1 expression also reduced progressive muscle degeneration and fibrosis in the diaphragm of aged mdx mice. FHL1 enhanced NFATc1 activation of the utrophin promoter and increased sarcolemmal expression of utrophin in muscles of mdx mice, directing the assembly of a substitute utrophin-glycoprotein complex, and revealing a novel FHL1-NFATc1-utrophin signaling axis that can functionally compensate for dystrophin.
Collapse
Affiliation(s)
- Colleen E D'Arcy
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, VIC 3800, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|