1
|
Furrer R, Dilbaz S, Steurer SA, Santos G, Karrer-Cardel B, Ritz D, Sinnreich M, Handschin C. Metabolic dysregulation contributes to the development of dysferlinopathy. Life Sci Alliance 2025; 8:e202402991. [PMID: 40021220 PMCID: PMC11871293 DOI: 10.26508/lsa.202402991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/03/2025] Open
Abstract
Dysferlin is a transmembrane protein that plays a prominent role in membrane repair of damaged muscle fibers. Accordingly, mutations in the dysferlin gene cause progressive muscular dystrophies, collectively referred to as dysferlinopathies for which no effective treatment exists. Unexpectedly, experimental approaches that successfully restore membrane repair fail to prevent a dystrophic phenotype, suggesting that additional, hitherto unknown dysferlin-dependent functions contribute to the development of the pathology. Our experiments revealed an altered metabolic phenotype in dysferlin-deficient muscles, characterized by (1) mitochondrial abnormalities and elevated death signaling and (2) increased glucose uptake, reduced glycolytic protein levels, and pronounced glycogen accumulation. Strikingly, elevating mitochondrial volume density and muscle glycogen accelerates disease progression; whereas, improvement of mitochondrial function and recruitment of muscle glycogen with exercise ameliorated functional parameters in a mouse model of dysferlinopathy. Collectively, our results not only shed light on a metabolic function of dysferlin but also imply new therapeutic avenues aimed at promoting mitochondrial function and normalizing muscle glycogen to ameliorate dysferlinopathies, complementing efforts that target membrane repair.
Collapse
Affiliation(s)
| | - Sedat Dilbaz
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - Gesa Santos
- Biozentrum, University of Basel, Basel, Switzerland
| | | | - Danilo Ritz
- Biozentrum, University of Basel, Basel, Switzerland
| | - Michael Sinnreich
- Department of Biomedicine and Neurology, University and University Hospital Basel, Basel, Switzerland
| | | |
Collapse
|
2
|
Kabir A, Mukilarasi B, Manohar A, Gadani M, Sinha AK, Sharma P, Verma A, Selvaraj V, Sudhakar S. Protein bioactive complexes promote osteogenesis under microgravity environment. Int J Biol Macromol 2025; 303:140483. [PMID: 39904451 DOI: 10.1016/j.ijbiomac.2025.140483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025]
Abstract
The space microgravity environment and cosmic radiation pose a significant threat to musculoskeletal health, particularly bone mass. However, the critical mechanism underlying space-induced bone loss and its relation to cellular oxidative stress remains unclear. Currently used bone-loss-reversing drugs face limitations like poor efficacy and metabolic defects. Herein, we revealed that simulated microgravity (SMG) induces reactive oxygen species (ROS), negatively impacting osteoblasts, causing cytoskeletal damage, and downregulating osteogenic genes. To combat this, we designed protein-zein nanocages loaded with a chimeric non-enzymatic cocktail (ZNAC) containing ascorbic acid, resveratrol, luteolin, coenzyme Q, and glutathione. These nanocages (~200 nm) demonstrated excellent stability, biocompatibility, and antioxidant properties compared to free drugs. We investigated the effects of ZNAC under SMG using two experimental models: MC3T3 pre-osteoblast/MG63 osteoblasts and regenerating zebrafish scales that represent compositional and physiological/pathophysiological analogy with mammalian system. ZNAC effectively reduced SMG-induced ROS, preserved cytoskeletal integrity, and enhanced alkaline phosphatase (ALP) activity along with the expression of osteogenic genes such as RUNX2 and Col1A1. In zebrafish scales, it increased osteogenic gene expression, calcification, and the calcium/phosphorus ratio, indicating enhanced scale regeneration. These findings suggest that ZNAC is a promising candidate for bone regeneration, offering potential solutions for maintaining astronaut health during extended space missions.
Collapse
Affiliation(s)
- Anisha Kabir
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - B Mukilarasi
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - Anagha Manohar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - Maulesh Gadani
- Space Applications Centre, Indian Space Research Organisation, Ahmedabad, Gujarat 380015, India.
| | - Anurag Kumar Sinha
- Human Space Flight Centre, Antariksh Bhavan, New BEL Road, Bengaluru 560 094, India.
| | - Payal Sharma
- Space Applications Centre, Indian Space Research Organisation, Ahmedabad, Gujarat 380015, India.
| | - Anurag Verma
- Space Applications Centre, Indian Space Research Organisation, Ahmedabad, Gujarat 380015, India.
| | - Vimalraj Selvaraj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - Swathi Sudhakar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| |
Collapse
|
3
|
Mackels L, Mariot V, Buscemi L, Servais L, Dumonceaux J. Impact of Disease Severity and Disease-Modifying Therapies on Myostatin Levels in SMA Patients. Int J Mol Sci 2024; 25:8763. [PMID: 39201450 PMCID: PMC11354404 DOI: 10.3390/ijms25168763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 08/02/2024] [Accepted: 08/06/2024] [Indexed: 09/02/2024] Open
Abstract
Clinical trials with treatments inhibiting myostatin pathways to increase muscle mass are currently ongoing in spinal muscular atrophy. Given evidence of potential myostatin pathway downregulation in Spinal Muscular Atrophy (SMA), restoring sufficient myostatin levels using disease-modifying treatments (DMTs) might arguably be necessary prior to considering myostatin inhibitors as an add-on treatment. This retrospective study assessed pre-treatment myostatin and follistatin levels' correlation with disease severity and explored their alteration by disease-modifying treatment in SMA. We retrospectively collected clinical characteristics, motor scores, and mysotatin and follistatin levels between 2018 and 2020 in 25 Belgian patients with SMA (SMA1 (n = 13), SMA2 (n = 6), SMA 3 (n = 6)) and treated by nusinersen. Data were collected prior to treatment and after 2, 6, 10, 18, and 30 months of treatment. Myostatin levels correlated with patients' age, weight, SMA type, and motor function before treatment initiation. After treatment, we observed correlations between myostatin levels and some motor function scores (i.e., MFM32, HFMSE, 6MWT), but no major effect of nusinersen on myostatin or follistatin levels over time. In conclusion, further research is needed to determine if DMTs can impact myostatin and follistatin levels in SMA, and how this could potentially influence patient selection for ongoing myostatin inhibitor trials.
Collapse
Affiliation(s)
- Laurane Mackels
- Adult Neurology Department, Citadelle Hospital, 1 Boulevard Du 12e De Ligne, 4000 Liege, Belgium
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK;
| | - Virginie Mariot
- NIHR Great Ormond Street Hospital Biomedical Research Centre and Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK;
| | - Laura Buscemi
- Neuromuscular Center, Citadelle Hospital, 1 Boulevard Du 12e De Ligne, 4000 Liege, Belgium;
| | - Laurent Servais
- MDUK Oxford Neuromuscular Center, Department of Paediatrics, NIHR Oxford Biomedical Research Centre, University of Oxford, Oxford OX3 9DU, UK;
- Neuromuscular Center, Division of Paediatrics, University Hospital of Liège, University of Liège, Boulevard Du 12e De Ligne, 4000 Liege, Belgium
| | - Julie Dumonceaux
- NIHR Great Ormond Street Hospital Biomedical Research Centre and Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK;
| |
Collapse
|
4
|
Khodabukus A, Prabhu NK, Roberts T, Buldo M, Detwiler A, Fralish ZD, Kondash ME, Truskey GA, Koves TR, Bursac N. Bioengineered Model of Human LGMD2B Skeletal Muscle Reveals Roles of Intracellular Calcium Overload in Contractile and Metabolic Dysfunction in Dysferlinopathy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2400188. [PMID: 38887849 PMCID: PMC11336985 DOI: 10.1002/advs.202400188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Revised: 03/19/2024] [Indexed: 06/20/2024]
Abstract
Dysferlin is a multi-functional protein that regulates membrane resealing, calcium homeostasis, and lipid metabolism in skeletal muscle. Genetic loss of dysferlin results in limb girdle muscular dystrophy 2B/2R (LGMD2B/2R) and other dysferlinopathies - rare untreatable muscle diseases that lead to permanent loss of ambulation in humans. The mild disease severity in dysferlin-deficient mice and diverse genotype-phenotype relationships in LGMD2B patients have prompted the development of new in vitro models for personalized studies of dysferlinopathy. Here the first 3-D tissue-engineered hiPSC-derived skeletal muscle ("myobundle") model of LGMD2B is described that exhibits compromised contractile function, calcium-handling, and membrane repair, and transcriptomic changes indicative of impaired oxidative metabolism and mitochondrial dysfunction. In response to the fatty acid (FA) challenge, LGMD2B myobundles display mitochondrial deficits and intracellular lipid droplet (LD) accumulation. Treatment with the ryanodine receptor (RyR) inhibitor dantrolene or the dissociative glucocorticoid vamorolone restores LGMD2B contractility, improves membrane repair, and reduces LD accumulation. Lastly, it is demonstrated that chemically induced chronic RyR leak in healthy myobundles phenocopies LGMD2B contractile and metabolic deficit, but not the loss of membrane repair capacity. Together, these results implicate intramyocellular Ca2+ leak as a critical driver of dysferlinopathic phenotype and validate the myobundle system as a platform to study LGMD2B pathogenesis.
Collapse
Affiliation(s)
| | - Neel K. Prabhu
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Taylor Roberts
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Meghan Buldo
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | - Amber Detwiler
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Megan E. Kondash
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| | | | - Timothy R. Koves
- Duke Molecular Physiology InstituteDuke UniversityDurhamNC27708USA
| | - Nenad Bursac
- Department of Biomedical EngineeringDuke UniversityDurhamNC27708USA
| |
Collapse
|
5
|
Gonzalez-Ponce F, Ramirez-Villafaña M, Gomez-Ramirez EE, Saldaña-Cruz AM, Gallardo-Moya SG, Rodriguez-Jimenez NA, Jacobo-Cuevas H, Nava-Valdivia CA, Avalos-Salgado FA, Totsuka-Sutto S, Cardona-Muñoz EG, Valdivia-Tangarife ER. Role of Myostatin in Rheumatoid Arthritis: A Review of the Clinical Impact. Diagnostics (Basel) 2024; 14:1085. [PMID: 38893612 PMCID: PMC11171688 DOI: 10.3390/diagnostics14111085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 06/21/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory disease that affects synovial joints and that frequently involves extra-articular organs. A multiplicity of interleukins (IL) participates in the pathogenesis of RA, including IL-6, IL-1β, transforming growth factor-beta (TGF-β), and tumor necrosis factor (TNF)-α; immune cells such as monocytes, T and B lymphocytes, and macrophages; and auto-antibodies, mainly rheumatoid factor and anti-citrullinated protein antibodies (ACPAs). Skeletal muscle is also involved in RA, with many patients developing muscle wasting and sarcopenia. Several mechanisms are involved in the myopenia observed in RA, and one of them includes the effects of some interleukins and myokines on myocytes. Myostatin is a myokine member of the TGF-β superfamily; the overproduction of myostatin acts as a negative regulator of growth and differentiates the muscle fibers, limiting their number and size. Recent studies have identified abnormalities in the serum myostatin levels of RA patients, and these have been found to be associated with muscle wasting and other manifestations of severe RA. This review analyzes recent information regarding the relationship between myostatin levels and clinical manifestations of RA and the relevance of myostatin as a therapeutic target for future research.
Collapse
Affiliation(s)
- Fabiola Gonzalez-Ponce
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (F.G.-P.); (M.R.-V.); (E.E.G.-R.); (A.M.S.-C.); (N.A.R.-J.); (S.T.-S.); (E.G.C.-M.)
| | - Melissa Ramirez-Villafaña
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (F.G.-P.); (M.R.-V.); (E.E.G.-R.); (A.M.S.-C.); (N.A.R.-J.); (S.T.-S.); (E.G.C.-M.)
| | - Eli Efrain Gomez-Ramirez
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (F.G.-P.); (M.R.-V.); (E.E.G.-R.); (A.M.S.-C.); (N.A.R.-J.); (S.T.-S.); (E.G.C.-M.)
| | - Ana Miriam Saldaña-Cruz
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (F.G.-P.); (M.R.-V.); (E.E.G.-R.); (A.M.S.-C.); (N.A.R.-J.); (S.T.-S.); (E.G.C.-M.)
| | - Sergio Gabriel Gallardo-Moya
- Programa de Doctorado en Farmacología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico; (S.G.G.-M.); (F.A.A.-S.)
| | - Norma Alejandra Rodriguez-Jimenez
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (F.G.-P.); (M.R.-V.); (E.E.G.-R.); (A.M.S.-C.); (N.A.R.-J.); (S.T.-S.); (E.G.C.-M.)
| | - Heriberto Jacobo-Cuevas
- Programa de Postdoctorado, Departamento de Psicología Básica, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Cesar Arturo Nava-Valdivia
- Departamento de Microbiología y Patología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico;
| | - Felipe Alexis Avalos-Salgado
- Programa de Doctorado en Farmacología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico; (S.G.G.-M.); (F.A.A.-S.)
| | - Sylvia Totsuka-Sutto
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (F.G.-P.); (M.R.-V.); (E.E.G.-R.); (A.M.S.-C.); (N.A.R.-J.); (S.T.-S.); (E.G.C.-M.)
| | - Ernesto German Cardona-Muñoz
- Instituto de Terapeutica Experimental y Clínica, Programa de Doctorado en Farmacología, Departamento de Fisiología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico; (F.G.-P.); (M.R.-V.); (E.E.G.-R.); (A.M.S.-C.); (N.A.R.-J.); (S.T.-S.); (E.G.C.-M.)
| | | |
Collapse
|
6
|
van de Velde NM, Koeks Z, Signorelli M, Verwey N, Overzier M, Bakker JA, Sajeev G, Signorovitch J, Ricotti V, Verschuuren J, Brown K, Spitali P, Niks EH. Longitudinal Assessment of Creatine Kinase, Creatine/Creatinine ratio, and Myostatin as Monitoring Biomarkers in Becker Muscular Dystrophy. Neurology 2023; 100:e975-e984. [PMID: 36849458 PMCID: PMC9990441 DOI: 10.1212/wnl.0000000000201609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 10/11/2022] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND AND OBJECTIVES The slow and variable disease progression of Becker muscular dystrophy (BMD) urges the development of biomarkers to facilitate clinical trials. We explored changes in 3 muscle-enriched biomarkers in serum of patients with BMD over 4-year time and studied associations with disease severity, disease progression, and dystrophin levels in BMD. METHODS We quantitatively measured creatine kinase (CK) using the International Federation of Clinical Chemistry reference method, creatine/creatinineratio (Cr/Crn) using liquid chromatography-tandem mass spectrometry, and myostatin with ELISA in serum and assessed functional performance using the North Star Ambulatory Assessment (NSAA), 10-meter run velocity (TMRv), 6-Minute Walking Test (6MWT), and forced vital capacity in a 4-year prospective natural history study. Dystrophin levels were quantified in the tibialis anterior muscle using capillary Western immunoassay. The correlation between biomarkers, age, functional performance, mean annual change, and prediction of concurrent functional performance was analyzed using linear mixed models. RESULTS Thirty-four patients with 106 visits were included. Eight patients were nonambulant at baseline. Cr/Crn and myostatin were highly patient specific (intraclass correlation coefficient for both = 0.960). Cr/Crn was strongly negatively correlated, whereas myostatin was strongly positively correlated with the NSAA, TMRv, and 6MWT (Cr/Crn rho = -0.869 to -0.801 and myostatin rho = 0.792 to 0.842, all p < 0.001). CK showed a negative association with age (p = 0.0002) but was not associated with patients' performance. Cr/Crn and myostatin correlated moderately with the average annual change of the 6MWT (rho = -0.532 and 0.555, p = 0.02). Dystrophin levels did not correlate with the selected biomarkers nor with performance. Cr/Crn, myostatin, and age could explain up to 75% of the variance of concurrent functional performance of the NSAA, TMRv, and 6MWT. DISCUSSION Both Cr/Crn and myostatin could potentially serve as monitoring biomarkers in BMD, as higher Cr/Crn and lower myostatin were associated with lower motor performance and predictive of concurrent functional performance when combined with age. Future studies are needed to more precisely determine the context of use of these biomarkers.
Collapse
Affiliation(s)
- Nienke M van de Velde
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Zaïda Koeks
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Mirko Signorelli
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Nisha Verwey
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Maurice Overzier
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Jaap A Bakker
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Gautam Sajeev
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - James Signorovitch
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Valeria Ricotti
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Jan Verschuuren
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Kristy Brown
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Pietro Spitali
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom
| | - Erik H Niks
- From the Departments of Neurology (N.M.V., Z.K., J.V., E.H.N.), Biomedical Data Sciences (M.S.), Human Genetics (N.V., M.O., P.S.), and Clinical Chemistry and Laboratory Medicine (J.A.B.), Leiden University Medical Center, the Netherlands; Duchenne Center Netherlands (N.M.V., J.V., P.S., E.H.N.); European Reference Network for Rare Neuromuscular Diseases [ERN EURO-NMD] (N.M.V., Z.K., N.V., M.O., J.V., P.S., E.H.N.); Mathematical Institute (M.S.), Leiden University, the Netherlands; Analysis Group Inc (G.S., J.S.), Boston, MA; Solid Biosciences Inc (V.R., K.B.), Cambridge, MA; and NIHR Great Ormond Street Hospital Biomedical Research Centre (V.R.), Great Ormond Street Institute of Child Health, University College London, & Great Ormond Street Hospital Trust, United Kingdom.
| |
Collapse
|
7
|
Moore U, Fernández-Simón E, Schiava M, Cox D, Gordish-Dressman H, James MK, Mayhew A, Wilson I, Guglieri M, Rufibach L, Blamire A, Carlier PG, Mori-Yoshimura M, Day JW, Jones KJ, Bharucha-Goebel DX, Salort-Campana E, Pestronk A, Walter MC, Paradas C, Stojkovic T, Bravver E, Pegoraro E, Mendell JR, Bushby K, Diaz-Manera J, Straub V. Myostatin and follistatin as monitoring and prognostic biomarkers in dysferlinopathy. Neuromuscul Disord 2023; 33:199-207. [PMID: 36689846 DOI: 10.1016/j.nmd.2023.01.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 12/15/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
Myostatin is a myokine which acts upon skeletal muscle to inhibit growth and regeneration. Myostatin is endogenously antagonised by follistatin. This study assessed serum myostatin and follistatin concentrations as monitoring or prognostic biomarkers in dysferlinopathy, an autosomal recessively inherited muscular dystrophy. Myostatin was quantified twice with a three-year interval in 76 patients with dysferlinopathy and 38 controls. Follistatin was quantified in 62 of these patients at the same timepoints, and in 31 controls. Correlations with motor function, muscle fat fraction and contractile cross-sectional area were performed. A regression model was used to account for confounding variables. Baseline myostatin, but not follistatin, correlated with baseline function and MRI measures. However, in individual patients, three-year change in myostatin did not correlate with functional or MRI changes. Linear modelling demonstrated that function, serum creatine kinase and C-reactive protein, but not age, were independently related to myostatin concentration. Baseline myostatin concentration predicted loss of ambulation but not rate of change of functional or MRI measures, even when relative inhibition with follistatin was considered. With adjustment for extra-muscular causes of variation, myostatin could form a surrogate measure of functional ability or muscle mass, however myostatin inhibition does not form a promising treatment target in dysferlinopathy.
Collapse
Affiliation(s)
- Ursula Moore
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Esther Fernández-Simón
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Marianela Schiava
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Dan Cox
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Heather Gordish-Dressman
- Center for Translational Science, Division of Biostatistics and Study Methodology, Children's National Health System, Washington, DC, USA; Pediatrics, Epidemiology and Biostatistics, George Washington University, Washington, DC, USA
| | - Meredith K James
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Anna Mayhew
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Ian Wilson
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Michela Guglieri
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | | | - Andrew Blamire
- Magnetic Resonance Centre, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | | | - Madoka Mori-Yoshimura
- Department of Neurology, National Center Hospital, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - John W Day
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristi J Jones
- The Children's Hospital at Westmead and The University of Sydney, Sydney, NSW, Australia
| | - Diana X Bharucha-Goebel
- Department of Neurology, Children's National Health System, Washington, DC, USA; National Institutes of Health (NINDS), Bethesda, MD, USA
| | | | - Alan Pestronk
- Department of Neurology, Washington University School of Medicine, St. Louis, MO, USA
| | - Maggie C Walter
- Friedrich-Baur-Institute, Department of Neurology, LudwigMaximilians-University of Munich, Munich, Germany
| | - Carmen Paradas
- Neuromuscular Unit, Department of Neurology, Hospital U. Virgen del Rocío/Instituto de Biomedicina de Sevilla, Sevilla, Spain
| | - Tanya Stojkovic
- Centre de reference des maladies neuromusculaires, Institut de Myologie, AP-HP, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, France
| | - Elena Bravver
- Neuroscience Institute, Carolinas Neuromuscular/ALS-MDA Center, Carolinas HealthCare System, Charlotte, NC, USA
| | - Elena Pegoraro
- Department of Neuroscience, University of Padova, Padua, Italy
| | - Jerry R Mendell
- The Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Kate Bushby
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Jordi Diaz-Manera
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK; Neuromuscular Disorders Unit, Neurology Department, Hospital de la Santa Creu I Sant Pau, Barcelona, Spain; Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Madrid, Spain
| | - Volker Straub
- The John Walton Muscular Dystrophy Research Centre, Translational and Clinical Research Institute, Newcastle University and Newcastle Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK.
| | | |
Collapse
|
8
|
Jin Z, Choe HM, Lv S, Chang S, Yin X. Esophageal striated muscle hypertrophy and muscle fiber type transformation in MSTN knockout pigs. Transgenic Res 2022; 31:341-349. [DOI: 10.1007/s11248-022-00309-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/18/2022] [Indexed: 11/28/2022]
|
9
|
Bijeh N, Mohammadnia-Ahmadi M, Hooshamnd-Moghadam B, Eskandari M, Golestani F. Effects of Soy Milk in Conjunction With Resistance Training on Physical Performance and Skeletal Muscle Regulatory Markers in Older Men. Biol Res Nurs 2022; 24:294-307. [PMID: 35332795 DOI: 10.1177/10998004211073123] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Purpose: We aimed to determine the effects of 12 weeks of soy milk consumption combined with resistance training (RT) on body composition, physical performance, and skeletal muscle regulatory markers in older men. Methods: In this randomized clinical trial study, 60 healthy elderly men (age = 65.63 ± 3.16 years) were randomly assigned to four groups: resistance training (RT; n = 15), soy milk consumption (SMC; n = 15), resistance training + soy milk (RSM; n = 15), and control (CON; n = 15) groups. The study was double-blind for the soy milk/placebo. Participants in RT and RSM groups performed resistance training (3 times/week) for 12 weeks. Participants in the SMC and RSM groups consumed 240 mL of soy milk daily. Body composition [body mass (BM), body fat percent (BFP), waist-hip ratio (WHR), and fat mass (FM)], physical performance [upper body strength (UBS), lower body strength (LBS), VO2max, upper anaerobic power, lower anaerobic power, and handgrip strength], and serum markers [follistatin, myostatin, myostatin-follistatin ratio (MFR), and growth and differentiation factor 11 (GDF11)] were evaluated before and after interventions. Results: All 3 interventions significantly (p < 0.05) increased serum follistatin concentrations (RT = 1.7%, SMC = 2.9%, RSM = 7.8%) and decreased serum myostatin (RT = -1.3% SMC = -5.4%, RSM = -0.5%) and GDF11 concentrations (RT = -1.4%, SMC = -1.4%, RSM = -9.0%), and MFR (RT = -2.6%, SMC = -3.2%, RSM = -12%). In addition, we observed significant reduction in all 3 intervention groups in BFP (RT = -3.6%, SMC = -1.4%, RSM = -6.0%), WHR (RT = -2.2%, SMC = -2.1%, RSM = -4.3%), and FM (RT = -9.6%, SMC = -3.8%, RSM = -11.0%). Moreover, results found significant increase only in RT and RSM groups for muscle mass (RT = 3.8% and RSM = 11.8%), UBS (RT = 10.9% and RSM = 21.8%), LBS (RT = 4.3% and RSM = 7.8%), upper anaerobic power (RT = 7.8% and RSM = 10.3%), and lower anaerobic power (RT = 4.6% and RSM = 8.9%). Handgrip strength were significantly increased in all 3 intervention groups (RT = 7.0%, SMC = 6.9%, RSM = 43.0%). VO2max significantly increased only in RSM (1.7%) after 12 weeks of intervention. Additionally, significant differences were observed between the changes for all variables in the RSM group compared to RT, SMC, and CON groups (p < 0.05). Conclusions: There were synergistic effects of soy milk and RT for skeletal muscle regulatory markers, body composition, and physical performance. Results of the present study support the importance of soy milk in conjunction with RT for older men.
Collapse
Affiliation(s)
- Nahid Bijeh
- Department of Exercise Physiology, 48440Ferdowsi University of Mashhad, Mashhad, Iran
| | | | | | - Mozhgan Eskandari
- Department of Exercise Physiology, 48528University of Birjand, Birjand, Iran
| | - Fateme Golestani
- Department of Exercise Physiology, 48528University of Birjand, Birjand, Iran
| |
Collapse
|
10
|
Demonbreun AR, Fallon KS, Oosterbaan CC, Vaught LA, Reiser NL, Bogdanovic E, Velez MP, Salamone IM, Page PGT, Hadhazy M, Quattrocelli M, Barefield DY, Wood LD, Gonzalez JP, Morris C, McNally EM. Anti-latent TGFβ binding protein 4 antibody improves muscle function and reduces muscle fibrosis in muscular dystrophy. Sci Transl Med 2021; 13:eabf0376. [PMID: 34516828 PMCID: PMC9559620 DOI: 10.1126/scitranslmed.abf0376] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Duchenne muscular dystrophy, like other muscular dystrophies, is a progressive disorder hallmarked by muscle degeneration, inflammation, and fibrosis. Latent transforming growth factor β (TGFβ) binding protein 4 (LTBP4) is an extracellular matrix protein found in muscle. LTBP4 sequesters and inhibits a precursor form of TGFβ. LTBP4 was originally identified from a genome-wide search for genetic modifiers of muscular dystrophy in mice, where there are two different alleles. The protective form of LTBP4, which contains an insertion of 12 amino acids in the protein’s hinge region, was linked to increased sequestration of latent TGFβ, enhanced muscle membrane stability, and reduced muscle fibrosis. The deleterious form of LTBP4 protein, lacking 12 amino acids, was more susceptible to proteolysis and promoted release of latent TGF-β, and together, these data underscored the functional role of LTBP4’s hinge. Here, we generated a monoclonal human anti-LTBP4 antibody directed toward LTBP4’s hinge region. In vitro, anti-LTBP4 bound LTBP4 protein and reduced LTBP4 proteolytic cleavage. In isolated myofibers, the LTBP4 antibody stabilized the sarcolemma from injury. In vivo, anti-LTBP4 treatment of dystrophic mice protected muscle against force loss induced by eccentric contraction. Anti-LTBP4 treatment also reduced muscle fibrosis and enhanced muscle force production, including in the diaphragm muscle, where respiratory function was improved. Moreover, the anti-LTBP4 in combination with prednisone, a standard of care for Duchenne muscular dystrophy, further enhanced muscle function and protected against injury in mdx mice. These data demonstrate the potential of anti-LTBP4 antibodies to treat muscular dystrophy.
Collapse
Affiliation(s)
- Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA.,Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - Katherine S Fallon
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Claire C Oosterbaan
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Lauren A Vaught
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Nina L Reiser
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Elena Bogdanovic
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Matthew P Velez
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Isabella M Salamone
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Patrick G T Page
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Mattia Quattrocelli
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA.,Department of Pharmacology, Northwestern University, Chicago, IL 60611, USA
| | - David Y Barefield
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| | | | | | | | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University, Chicago, IL 60611, USA
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Recent terminations of clinical trials of myostatin inhibitors in muscular dystrophy have raised questions about the predictiveness of mouse models for this therapeutic strategy. RECENT FINDINGS A variety of myostatin inhibitors have been developed for preclinical and clinical studies. These inhibitors have ameliorated the phenotype of many but not all mouse models of muscular dystrophy. However, randomized double-blinded placebo controlled trials in both pediatric and adult muscular dystrophies have, as of yet, not demonstrated functional improvement. SUMMARY The present article will review the preclinical promise of myostatin inhibitors, the clinical trial experience to date of these inhibitors in muscular dystrophy, and the potential reasons for the lack of observed translation.
Collapse
|
12
|
Antimyostatin Treatment in Health and Disease: The Story of Great Expectations and Limited Success. Cells 2021; 10:cells10030533. [PMID: 33802348 PMCID: PMC8001237 DOI: 10.3390/cells10030533] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/14/2022] Open
Abstract
In the past 20 years, myostatin, a negative regulator of muscle mass, has attracted attention as a potential therapeutic target in muscular dystrophies and other conditions. Preclinical studies have shown potential for increasing muscular mass and ameliorating the pathological features of dystrophic muscle by the inhibition of myostatin in various ways. However, hardly any clinical trials have proven to translate the promising results from the animal models into patient populations. We present the background for myostatin regulation, clinical and preclinical results and discuss why translation from animal models to patients is difficult. Based on this, we put the clinical relevance of future antimyostatin treatment into perspective.
Collapse
|
13
|
Huot JR, Pin F, Narasimhan A, Novinger LJ, Keith AS, Zimmers TA, Willis MS, Bonetto A. ACVR2B antagonism as a countermeasure to multi-organ perturbations in metastatic colorectal cancer cachexia. J Cachexia Sarcopenia Muscle 2020; 11:1779-1798. [PMID: 33200567 PMCID: PMC7749603 DOI: 10.1002/jcsm.12642] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 09/11/2020] [Accepted: 10/12/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Advanced colorectal cancer (CRC) is often accompanied by the development of liver metastases, as well as cachexia, a multi-organ co-morbidity primarily affecting skeletal (SKM) and cardiac muscles. Activin receptor type 2B (ACVR2B) signalling is known to cause SKM wasting, and its inhibition restores SKM mass and prolongs survival in cancer. Using a recently generated mouse model, here we tested whether ACVR2B blockade could preserve multiple organs, including skeletal and cardiac muscle, in the presence of metastatic CRC. METHODS NSG male mice (8 weeks old) were injected intrasplenically with HCT116 human CRC cells (mHCT116), while sham-operated animals received saline (n = 5-10 per group). Sham and tumour-bearing mice received weekly injections of ACVR2B/Fc, a synthetic peptide inhibitor of ACVR2B. RESULTS mHCT116 hosts displayed losses in fat mass ( - 79%, P < 0.0001), bone mass ( - 39%, P < 0.05), and SKM mass (quadriceps: - 22%, P < 0.001), in line with reduced muscle cross-sectional area ( - 24%, P < 0.01) and plantarflexion force ( - 28%, P < 0.05). Further, despite only moderately affected heart size, cardiac function was significantly impaired (ejection fraction %: - 16%, P < 0.0001; fractional shortening %: - 25%, P < 0.0001) in the mHCT116 hosts. Conversely, ACVR2B/Fc preserved fat mass ( + 238%, P < 0.001), bone mass ( + 124%, P < 0.0001), SKM mass (quadriceps: + 31%, P < 0.0001), size (cross-sectional area: + 43%, P < 0.0001) and plantarflexion force ( + 28%, P < 0.05) in tumour hosts. Cardiac function was also completely preserved in tumour hosts receiving ACVR2B/Fc (ejection fraction %: + 19%, P < 0.0001), despite no effect on heart size. RNA sequencing analysis of heart muscle revealed rescue of genes related to cardiac development and contraction in tumour hosts treated with ACVR2B/Fc. CONCLUSIONS Our metastatic CRC model recapitulates the multi-systemic derangements of cachexia by displaying loss of fat, bone, and SKM along with decreased muscle strength in mHCT116 hosts. Additionally, with evidence of severe cardiac dysfunction, our data support the development of cardiac cachexia in the occurrence of metastatic CRC. Notably, ACVR2B antagonism preserved adipose tissue, bone, and SKM, whereas muscle and cardiac functions were completely maintained upon treatment. Altogether, our observations implicate ACVR2B signalling in the development of multi-organ perturbations in metastatic CRC and further dictate that ACVR2B represents a promising therapeutic target to preserve body composition and functionality in cancer cachexia.
Collapse
Affiliation(s)
- Joshua R Huot
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fabrizio Pin
- Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ashok Narasimhan
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Leah J Novinger
- Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Teresa A Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.,Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.,Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Anatomy, Cell Biology and Physiology, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Otolaryngology-Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, USA.,Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN, USA
| |
Collapse
|
14
|
Shirvani H, Rahmati-Ahmadabad S, Kowsari E, Fry H, Kazemi M, Kaviani M. Effects of 2-week HMB-FA supplementation with or without eccentric resistance exercise on expression of some genes related to muscle protein turnover and serum irisin and IGF-1 concentrations. Gene 2020; 760:145018. [PMID: 32758580 DOI: 10.1016/j.gene.2020.145018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/06/2020] [Accepted: 07/30/2020] [Indexed: 02/05/2023]
Abstract
Protein turnover is a process that is regulated by several factors and can lead to muscle hypertrophy or atrophy. The purpose of the present study was to determine the effects of β-hydroxy-β-methylbutyrate free acid (HMB-FA) and eccentric resistance exercise on variables related to protein turnover in rats. Thirty-two male rats were randomly assigned into four groups of eight, including control, control-HMB, exercise, and exercise-HMB. Animals in HMB groups received 340 mg/kg/day for two weeks. Animals in the exercise groups performed one session of eccentric resistance exercise consisting of eight repetitions descending from a ladder with a slope of 80 degree, with an extra load of two times body weight (100% 1RM). Twenty-four hours after the exercise session, triceps brachii muscle and serum were collected for further analysis. Exercise and HMB-FA induced lower muscle myostatin and higher muscle Fibronectin type III domain containing 5 (FNDC5), P70-S6 kinase 1 gene expression, as well as higher serum irisin and IGF-1 concentrations. Exercise alone induced higher caspase-3 and caspase-8 gene expression while HMB-FA alone induced lower caspase 3 gene expression. HMB-FA supplement increased the effect of exercise on muscle FNDC5, myostatin, and P70-S6 kinase 1 gene expression. The interaction of exercise and HMBFA resulted in an additive effect, increasing serum irisin and IGF-1 concentrations. In conclusion, a 2-week HMB-FA supplementation paired with acute eccentric resistance exercise can positively affect some genes related to muscle protein turnover.
Collapse
Affiliation(s)
- Hossein Shirvani
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | | | - Elias Kowsari
- Exercise Physiology Research Center, Life Style Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Hillary Fry
- Division of Nutritional Sciences, Human Metabolic Research Unit, Cornell University, Ithaca, NY, USA
| | - Maryam Kazemi
- Division of Nutritional Sciences, Human Metabolic Research Unit, Cornell University, Ithaca, NY, USA
| | - Mojtaba Kaviani
- School of Nutrition and Dietetics, Acadia University, Wolfville, Nova Scotia, Canada.
| |
Collapse
|
15
|
Kramerova I, Marinov M, Owens J, Lee SJ, Becerra D, Spencer MJ. Myostatin inhibition promotes fast fibre hypertrophy but causes loss of AMP-activated protein kinase signalling and poor exercise tolerance in a model of limb-girdle muscular dystrophy R1/2A. J Physiol 2020; 598:3927-3939. [PMID: 33460149 DOI: 10.1113/jp279943] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/08/2020] [Indexed: 05/31/2024] Open
Abstract
KEY POINTS Limb-girdle muscular dystrophy R1 (LGMD R1) is caused by mutations in the CAPN3 gene and is characterized by progressive muscle loss, impaired mitochondrial function and reductions in the slow oxidative gene expression programme. Myostatin is a negative regulator of muscle growth, and its inhibition improves the phenotype in several muscle wasting disorders. The effect of genetic and pharmacological inhibition of myostatin signalling on the disease phenotype in a mouse model of LGMD R1 (CAPN3 knockout mouse-C3KO) was studied. Inhibition of myostatin signalling in C3KO muscles resulted in significant muscle hypertrophy; however, there were no improvements in muscle strength and exacerbation of exercise intolerance concomitant with further reduction of muscle oxidative capacity was observed. Inhibition of myostatin signalling is unlikely to be a valid therapeutic strategy for LGMD R1. ABSTRACT Limb-girdle muscular dystrophy R1 (LGMD R1) is caused by mutations in the CAPN3 gene and is characterized by progressive muscle loss, impaired mitochondrial function and reductions in the slow oxidative gene expression programme. There are currently no therapies available to patients. We sought to determine if induction of muscle growth, through myostatin inhibition, represents a viable therapeutic strategy for this disease. Myostatin is a negative regulator of muscle growth, and its inhibition improves the phenotype in several muscle wasting disorders. However, the effect of myostatin depends on the genetic and pathophysiological context and may not be efficacious in all contexts. We found that genetic inhibition of myostatin through overexpression of follistatin (an endogenous inhibitor of myostatin) in our LGMD R1 model (C3KO) resulted in 1.5- to 2-fold increase of muscle mass for the majority of limb muscles. However, muscle strength was not improved and exercise intolerance was exacerbated. Pharmacological inhibition of myostatin, using an anti-myostatin antibody, resulted in statistically significant increases in muscle mass; however, functional testing did not reveal changes in muscle strength nor endurance in treated C3KO mice. Histochemical and biochemical evaluation of follistatin overexpressing mice revealed a reduction in the percentage of oxidative fibres and decreased activation of AMP-activated protein kinase signalling in transgenics compared to C3KO muscles. Our data suggest that muscle hypertrophy, induced by myostatin inhibition, leads to loss of oxidative capacity, which further compromises metabolically impaired C3KO muscles and thus is unlikely to be a valid strategy for treatment of LGMD R1.
Collapse
Affiliation(s)
- Irina Kramerova
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Masha Marinov
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | | | - Se-Jin Lee
- The Jackson Laboratory and University of Connecticut School of Medicine, Farmington, CT, USA
| | - Diana Becerra
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Melissa J Spencer
- Department of Neurology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
16
|
Biglari S, Afousi AG, Mafi F, Shabkhiz F. High-intensity interval training-induced hypertrophy in gastrocnemius muscle via improved IGF-I/Akt/FoxO and myostatin/Smad signaling pathways in rats. Physiol Int 2020; 107:220-230. [PMID: 32644938 DOI: 10.1556/2060.2020.00020] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 04/04/2020] [Indexed: 12/17/2022]
Abstract
Objective It has been shown that high-intensity interval training (HIIT) leads to skeletal muscle hypertrophy; however, its mechanisms of cellular and molecular regulation are still unclear. The purpose of this study was to investigate the effect of HIIT on muscle hypertrophy and major signal transduction pathways. Design 12 male rats were randomly divided into two groups: control and HIIT. The exercise group performed 30-min HIIT in each session (5 × 4-min intervals running at 85-95% VO2max separated by 2-min active rest at 55-60% VO2max), 3 days/week for 8 weeks. Muscle fiber cross-sectional area (CSA) and the expression of signal transduction pathway proteins were determined in the gastrocnemius muscle. Results In the HIIT group, the expression of IGF-I, IGF-IR Akt, p-Akt, AMPKα, p-AMPKα and follistatin increased significantly, whereas a significant decrease was observed in the expression of FoxO1, p-FoxO1, myostatin, ActRIIB, Smad2/3 and p-Smad2/3 (P < 0.05). However, there were no significant differences between the HIIT and control groups in the expression of mTOR, p-mTOR, P70S6K, and p-P70S6K (P > 0.05). In addition, CSA and gastrocnemius muscle weight increased significantly in the HIIT group (P < 0.05). Conclusions HIIT induced muscle hypertrophy by improving IGF-I/Akt/FoxO and myostatin/Smad signal transduction pathways.
Collapse
Affiliation(s)
- Soheil Biglari
- Department of Exercise Physiology, Faculty of Physical Education and Exercise Sciences, University of Tehran, Tehran, Islamic Republic of Iran
| | - Alireza Ghardashi Afousi
- Department of Exercise Physiology, Faculty of Physical Education and Exercise Sciences, University of Tehran, Tehran, Islamic Republic of Iran
| | - Farnoosh Mafi
- Department of Exercise Physiology, Faculty of Physical Education and Exercise Sciences, University of Tehran, Tehran, Islamic Republic of Iran
| | - Fatemeh Shabkhiz
- Department of Exercise Physiology, Faculty of Physical Education and Exercise Sciences, University of Tehran, Tehran, Islamic Republic of Iran
| |
Collapse
|
17
|
Bagheri R, Moghadam BH, Church DD, Tinsley GM, Eskandari M, Moghadam BH, Motevalli MS, Baker JS, Robergs RA, Wong A. The effects of concurrent training order on body composition and serum concentrations of follistatin, myostatin and GDF11 in sarcopenic elderly men. Exp Gerontol 2020; 133:110869. [DOI: 10.1016/j.exger.2020.110869] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Revised: 01/14/2020] [Accepted: 02/05/2020] [Indexed: 01/18/2023]
|
18
|
Antunes-Correa LM, Trevizan PF, Bacurau AVN, Ferreira-Santos L, Gomes JLP, Urias U, Oliveira PA, Alves MJNN, de Almeida DR, Brum PC, Oliveira EM, Hajjar L, Kalil Filho R, Negrão CE. Effects of aerobic and inspiratory training on skeletal muscle microRNA-1 and downstream-associated pathways in patients with heart failure. J Cachexia Sarcopenia Muscle 2020; 11:89-102. [PMID: 31743617 PMCID: PMC7015255 DOI: 10.1002/jcsm.12495] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Revised: 07/26/2019] [Accepted: 08/12/2019] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The exercise intolerance in chronic heart failure with reduced ejection fraction (HFrEF) is mostly attributed to alterations in skeletal muscle. However, the mechanisms underlying the skeletal myopathy in patients with HFrEF are not completely understood. We hypothesized that (i) aerobic exercise training (AET) and inspiratory muscle training (IMT) would change skeletal muscle microRNA-1 expression and downstream-associated pathways in patients with HFrEF and (ii) AET and IMT would increase leg blood flow (LBF), functional capacity, and quality of life in these patients. METHODS Patients age 35 to 70 years, left ventricular ejection fraction (LVEF) ≤40%, New York Heart Association functional classes II-III, were randomized into control, IMT, and AET groups. Skeletal muscle changes were examined by vastus lateralis biopsy. LBF was measured by venous occlusion plethysmography, functional capacity by cardiopulmonary exercise test, and quality of life by Minnesota Living with Heart Failure Questionnaire. All patients were evaluated at baseline and after 4 months. RESULTS Thirty-three patients finished the study protocol: control (n = 10; LVEF = 25 ± 1%; six males), IMT (n = 11; LVEF = 31 ± 2%; three males), and AET (n = 12; LVEF = 26 ± 2%; seven males). AET, but not IMT, increased the expression of microRNA-1 (P = 0.02; percent changes = 53 ± 17%), decreased the expression of PTEN (P = 0.003; percent changes = -15 ± 0.03%), and tended to increase the p-AKTser473 /AKT ratio (P = 0.06). In addition, AET decreased HDAC4 expression (P = 0.03; percent changes = -40 ± 19%) and upregulated follistatin (P = 0.01; percent changes = 174 ± 58%), MEF2C (P = 0.05; percent changes = 34 ± 15%), and MyoD expression (P = 0.05; percent changes = 47 ± 18%). AET also increased muscle cross-sectional area (P = 0.01). AET and IMT increased LBF, functional capacity, and quality of life. Further analyses showed a significant correlation between percent changes in microRNA-1 and percent changes in follistatin mRNA (P = 0.001, rho = 0.58) and between percent changes in follistatin mRNA and percent changes in peak VO2 (P = 0.004, rho = 0.51). CONCLUSIONS AET upregulates microRNA-1 levels and decreases the protein expression of PTEN, which reduces the inhibitory action on the PI3K-AKT pathway that regulates the skeletal muscle tropism. The increased levels of microRNA-1 also decreased HDAC4 and increased MEF2c, MyoD, and follistatin expression, improving skeletal muscle regeneration. These changes associated with the increase in muscle cross-sectional area and LBF contribute to the attenuation in skeletal myopathy, and the improvement in functional capacity and quality of life in patients with HFrEF. IMT caused no changes in microRNA-1 and in the downstream-associated pathway. The increased functional capacity provoked by IMT seems to be associated with amelioration in the respiratory function instead of changes in skeletal muscle. ClinicalTrials.gov (Identifier: NCT01747395).
Collapse
Affiliation(s)
- Ligia M Antunes-Correa
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil.,School of Physical Education, University of Campinas (UNICAMP), Campinas, Brazil
| | - Patricia F Trevizan
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Aline V N Bacurau
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | | | - João L P Gomes
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Ursula Urias
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Patricia A Oliveira
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | | | - Dirceu R de Almeida
- Division of Cardiology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Patricia C Brum
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Edilamar M Oliveira
- School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| | - Ludhmila Hajjar
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Roberto Kalil Filho
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil
| | - Carlos Eduardo Negrão
- Heart Institute (InCor), University of São Paulo Medical School, São Paulo, Brazil.,School of Physical Education and Sport, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Mukund K, Subramaniam S. Skeletal muscle: A review of molecular structure and function, in health and disease. WILEY INTERDISCIPLINARY REVIEWS. SYSTEMS BIOLOGY AND MEDICINE 2020; 12:e1462. [PMID: 31407867 PMCID: PMC6916202 DOI: 10.1002/wsbm.1462] [Citation(s) in RCA: 283] [Impact Index Per Article: 56.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 07/03/2019] [Accepted: 07/03/2019] [Indexed: 12/11/2022]
Abstract
Decades of research in skeletal muscle physiology have provided multiscale insights into the structural and functional complexity of this important anatomical tissue, designed to accomplish the task of generating contraction, force and movement. Skeletal muscle can be viewed as a biomechanical device with various interacting components including the autonomic nerves for impulse transmission, vasculature for efficient oxygenation, and embedded regulatory and metabolic machinery for maintaining cellular homeostasis. The "omics" revolution has propelled a new era in muscle research, allowing us to discern minute details of molecular cross-talk required for effective coordination between the myriad interacting components for efficient muscle function. The objective of this review is to provide a systems-level, comprehensive mapping the molecular mechanisms underlying skeletal muscle structure and function, in health and disease. We begin this review with a focus on molecular mechanisms underlying muscle tissue development (myogenesis), with an emphasis on satellite cells and muscle regeneration. We next review the molecular structure and mechanisms underlying the many structural components of the muscle: neuromuscular junction, sarcomere, cytoskeleton, extracellular matrix, and vasculature surrounding muscle. We highlight aberrant molecular mechanisms and their possible clinical or pathophysiological relevance. We particularly emphasize the impact of environmental stressors (inflammation and oxidative stress) in contributing to muscle pathophysiology including atrophy, hypertrophy, and fibrosis. This article is categorized under: Physiology > Mammalian Physiology in Health and Disease Developmental Biology > Developmental Processes in Health and Disease Models of Systems Properties and Processes > Cellular Models.
Collapse
Affiliation(s)
- Kavitha Mukund
- Department of BioengineeringUniversity of CaliforniaSan DiegoCalifornia
| | - Shankar Subramaniam
- Department of Bioengineering, Bioinformatics & Systems BiologyUniversity of CaliforniaSan DiegoCalifornia
- Department of Computer Science and EngineeringUniversity of CaliforniaSan DiegoCalifornia
- Department of Cellular and Molecular Medicine and NanoengineeringUniversity of CaliforniaSan DiegoCalifornia
| |
Collapse
|
20
|
Guha TK, Pichavant C, Calos MP. Plasmid-Mediated Gene Therapy in Mouse Models of Limb Girdle Muscular Dystrophy. Mol Ther Methods Clin Dev 2019; 15:294-304. [PMID: 31890729 PMCID: PMC6923511 DOI: 10.1016/j.omtm.2019.10.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Accepted: 10/09/2019] [Indexed: 11/30/2022]
Abstract
We delivered plasmid DNA encoding therapeutic genes to the muscles of mouse models of limb girdle muscular dystrophy (LGMD) 2A, 2B, and 2D, deficient in calpain3, dysferlin, and alpha-sarcoglycan, respectively. We also delivered the human follistatin gene, which has the potential to increase therapeutic benefit. After intramuscular injection of DNA, electroporation was applied to enhance delivery to muscle fibers. When plasmids encoding the human calpain3 or dysferlin cDNA sequences were injected into quadriceps muscles of LGMD2A and LGMD2B mouse models, respectively, in 3-month studies, robust levels of calpain3 and dysferlin proteins were detected. We observed a statistically significant decrease in Evans blue dye penetration in LGMD2B mouse muscles after delivery of the dysferlin gene, consistent with repair of the muscle membrane defect in these mice. The therapeutic value of delivery of the genes for alpha-sarcoglycan and follistatin was documented by significant drops in Evans blue dye penetration in gastrocnemius muscles of LGMD2D mice. These results indicated for the first time that a combined gene therapy involving both alpha-sarcoglycan and follistatin would be valuable for LGMD2D patients. We suggest that this non-viral gene delivery method should be explored for its translational potential in patients.
Collapse
Affiliation(s)
- Tuhin K. Guha
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | - Christophe Pichavant
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | - Michele P. Calos
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| |
Collapse
|
21
|
Barton ER, Pham J, Brisson BK, Park S, Smith LR, Liu M, Tian Z, Hammers DW, Vassilakos G, Sweeney HL. Functional muscle hypertrophy by increased insulin-like growth factor 1 does not require dysferlin. Muscle Nerve 2019; 60:464-473. [PMID: 31323135 PMCID: PMC6771521 DOI: 10.1002/mus.26641] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 07/11/2019] [Accepted: 07/15/2019] [Indexed: 11/24/2022]
Abstract
INTRODUCTION Dysferlin loss-of-function mutations cause muscular dystrophy, accompanied by impaired membrane repair and muscle weakness. Growth promoting strategies including insulin-like growth factor 1 (IGF-1) could provide benefit but may cause strength loss or be ineffective. The objective of this study was to determine whether locally increased IGF-1 promotes functional muscle hypertrophy in dysferlin-null (Dysf-/- ) mice. METHODS Muscle-specific transgenic expression and postnatal viral delivery of Igf1 were used in Dysf-/- and control mice. Increased IGF-1 levels were confirmed by enzyme-linked immunosorbent assay. Testing for skeletal muscle mass and function was performed in male and female mice. RESULTS Muscle hypertrophy occurred in response to increased IGF-1 in mice with and without dysferlin. Male mice showed a more robust response compared with females. Increased IGF-1 did not cause loss of force per cross-sectional area in Dysf-/- muscles. DISCUSSION We conclude that increased local IGF-1 promotes functional hypertrophy when dysferlin is absent and reestablishes IGF-1 as a potential therapeutic for dysferlinopathies.
Collapse
Affiliation(s)
- Elisabeth R. Barton
- Anatomy and Cell Biology, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
- Applied Physiology and KinesiologyCollege of Health and Human Performance, University of FloridaGainesvilleFlorida
| | - Jennifer Pham
- Department of Physiology, Perleman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvania
| | - Becky K. Brisson
- Anatomy and Cell Biology, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - SooHyun Park
- Anatomy and Cell Biology, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Lucas R. Smith
- Anatomy and Cell Biology, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - Min Liu
- Department of Physiology, Perleman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvania
| | - Zuozhen Tian
- Anatomy and Cell Biology, School of Dental MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvania
| | - David W. Hammers
- Department of Pharmacology and Therapeutics, College of Medicine, University of FloridaGainesvilleFlorida
| | - Georgios Vassilakos
- Applied Physiology and KinesiologyCollege of Health and Human Performance, University of FloridaGainesvilleFlorida
| | - H. Lee Sweeney
- Department of Physiology, Perleman School of Medicine, University of PennsylvaniaPhiladelphiaPennsylvania
- Department of Pharmacology and Therapeutics, College of Medicine, University of FloridaGainesvilleFlorida
| |
Collapse
|
22
|
O'Connell TM, Pin F, Couch ME, Bonetto A. Treatment with Soluble Activin Receptor Type IIB Alters Metabolic Response in Chemotherapy-Induced Cachexia. Cancers (Basel) 2019; 11:cancers11091222. [PMID: 31438622 PMCID: PMC6770556 DOI: 10.3390/cancers11091222] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Revised: 08/07/2019] [Accepted: 08/16/2019] [Indexed: 02/08/2023] Open
Abstract
Some chemotherapeutic agents have been shown to lead to the severe wasting syndrome known as cachexia resulting in dramatic losses of both skeletal muscle and adipose tissue. Previous studies have shown that chemotherapy-induced cachexia is characterized by unique metabolic alterations. Recent results from our laboratory and others have shown that the use of ACVR2B/Fc, a soluble form of the activin receptor 2B (ACVR2B), can mitigate muscle wasting induced by chemotherapy, although the underlying mechanisms responsible for such protective effects are unclear. In order to understand the biochemical mechanisms through which ACVR2B/Fc functions, we employed a comprehensive, multi-platform metabolomics approach. Using both nuclear magnetic resonance (NMR) and mass-spectrometry (MS), we profiled the metabolome of both serum and muscle tissue from four groups of mice including (1) vehicle, (2) the chemotherapeutic agent, Folfiri, (3) ACVR2B/Fc alone, and (4) combined treatment with both Folfiri and ACVR2B/Fc. The metabolic profiles demonstrated large effects with Folfiri treatment and much weaker effects with ACVR2B/Fc treatment. Interestingly, a number of significant effects were observed in the co-treatment group, with the addition of ACVR2B/Fc providing some level of rescue to the perturbations induced by Folfiri alone. The most prominent of these were a normalization of systemic glucose and lipid metabolism. Identification of these pathways provides important insights into the mechanism by which ACVR2B/Fc protects against chemotherapy-induced cachexia.
Collapse
Affiliation(s)
- Thomas M O'Connell
- Department of Otolaryngology-Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Fabrizio Pin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Marion E Couch
- Department of Otolaryngology-Head & Neck Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
23
|
Significant body mass increase by oral administration of a cascade of shIL21-MSTN yeast-based DNA vaccine in mice. Biomed Pharmacother 2019; 118:109147. [PMID: 31302418 DOI: 10.1016/j.biopha.2019.109147] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 06/12/2019] [Accepted: 06/17/2019] [Indexed: 12/31/2022] Open
Abstract
Base on the practical of MSTN-specific yeast-based protein vaccine in mice as described previously, this research was designed for developing a better DNA vaccine (a cascade of shIL21-MSTN yeast-based DNA vaccine) than solely MSTN yeast-based DNA vaccine to block the endogenous MSTN in the murine model. We first constructed the target vectors, including CMV-driven MSTN expression vector and a combined shIL21-MSTN vector which containing MSTN expression cassette and shIL21 (short hairpin RNA-IL21) expression cassette. After necessary validation, recombinant yeast vaccines harboring different vectors were well prepared. Subsequently, after 2-month administration, the MSTN-specific immune response was detected with western blots. The commercial ELISA assays indicated that the production of IL21 and IL6 were decreased compared with control groups. More importantly, the MSTN-specific antibody titer was much higher in the shIL21-MSTN group than MSTN group, which was consistent with the western blots result. The most important finding was significant body mass increased after oral administration of these yeast-based DNA vaccines, in which the shIL21-MSTN vaccine is slightly higher than the sole MSTN vaccine in mice. In this study, we confirmed the role of different MSTN-specific yeast-based DNA vaccines on increasing body mass in mice, to provide a good inspiration for livestock breeding through the new type of immunoregulatory method. On the other hand, we also detected the possible modulating role of shIL21 on the dendritic cell-mediated immune function which needs more practical application and deeper exploration.
Collapse
|
24
|
Vo AH, Swaggart KA, Woo A, Gao QQ, Demonbreun AR, Fallon KS, Quattrocelli M, Hadhazy M, Page PGT, Chen Z, Eskin A, Squire K, Nelson SF, McNally EM. Dusp6 is a genetic modifier of growth through enhanced ERK activity. Hum Mol Genet 2019; 28:279-289. [PMID: 30289454 DOI: 10.1093/hmg/ddy349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 09/26/2018] [Indexed: 12/21/2022] Open
Abstract
Like other single-gene disorders, muscular dystrophy displays a range of phenotypic heterogeneity even with the same primary mutation. Identifying genetic modifiers capable of altering the course of muscular dystrophy is one approach to deciphering gene-gene interactions that can be exploited for therapy development. To this end, we used an intercross strategy in mice to map modifiers of muscular dystrophy. We interrogated genes of interest in an interval on mouse chromosome 10 associated with body mass in muscular dystrophy as skeletal muscle contributes significantly to total body mass. Using whole-genome sequencing of the two parental mouse strains combined with deep RNA sequencing, we identified the Met62Ile substitution in the dual-specificity phosphatase 6 (Dusp6) gene from the DBA/2 J (D2) mouse strain. DUSP6 is a broadly expressed dual-specificity phosphatase protein, which binds and dephosphorylates extracellular-signal-regulated kinase (ERK), leading to decreased ERK activity. We found that the Met62Ile substitution reduced the interaction between DUSP6 and ERK resulting in increased ERK phosphorylation and ERK activity. In dystrophic muscle, DUSP6 Met62Ile is strongly upregulated to counteract its reduced activity. We found that myoblasts from the D2 background were insensitive to a specific small molecule inhibitor of DUSP6, while myoblasts expressing the canonical DUSP6 displayed enhanced proliferation after exposure to DUSP6 inhibition. These data identify DUSP6 as an important regulator of ERK activity in the setting of muscle growth and muscular dystrophy.
Collapse
Affiliation(s)
- Andy H Vo
- Committee on Development, Regeneration and Stem Cell Biology, The University of Chicago, Chicago, IL
| | | | - Anna Woo
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Quan Q Gao
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Katherine S Fallon
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Mattia Quattrocelli
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Michele Hadhazy
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Patrick G T Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| | - Zugen Chen
- Departments of Human Genetics and Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ascia Eskin
- Departments of Human Genetics and Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kevin Squire
- Departments of Human Genetics and Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Stanley F Nelson
- Departments of Human Genetics and Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago IL
| |
Collapse
|
25
|
Negaresh R, Ranjbar R, Baker JS, Habibi A, Mokhtarzade M, Gharibvand MM, Fokin A. Skeletal Muscle Hypertrophy, Insulin-like Growth Factor 1, Myostatin and Follistatin in Healthy and Sarcopenic Elderly Men: The Effect of Whole-body Resistance Training. Int J Prev Med 2019; 10:29. [PMID: 30967915 PMCID: PMC6425763 DOI: 10.4103/ijpvm.ijpvm_310_17] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 12/05/2017] [Indexed: 01/12/2023] Open
Abstract
Background: Sarcopenia describes the inevitable deterioration in muscle mass and strength that accompanies biological aging. The purpose of this study was to investigate the effects of resistance training (RT) on quadriceps hypertrophy and related biochemistry in sarcopenic and healthy elderly men. Methods: A total of 31 elderly men (55–70 years old) were classified as sarcopenic and nonsarcopenic and were divided into two groups. Both groups participated in a progressive RT program for 8 weeks. Results: Data indicated that the strength in the sarcopenic group increased more than the healthy group (P < 0.05). Quadriceps cross-sectional area also increased more in the healthy group (P < 0.05). Myostatin concentration decreased in both groups after training (P < 0.05). Follistatin and testosterone increased in the healthy group; in contrast, only testosterone increased in the sarcopenic group after training (P < 0.05). Conclusions: The findings from this study suggest that RT improves muscle cross-sectional area and biomarker-related muscle loss in both healthy and sarcopenic elderly men. The findings also demonstrate that growth factor profiles at baseline and changes in testosterone levels play an important role in muscle hypertrophy observed in both groups.
Collapse
Affiliation(s)
- Raoof Negaresh
- Department of Sport Physiology, Shahid Chamran University, Ahvaz, Iran.,Department of Sport Physiology, Tarbiat Modares University, Tehran, Iran
| | - Rouholah Ranjbar
- Department of Sport Physiology, Shahid Chamran University, Ahvaz, Iran
| | - Julien S Baker
- School of Science and Sport, University of the West of Scotland, Paisley, Scotland
| | - Abdolhamid Habibi
- Department of Sport Physiology, Shahid Chamran University, Ahvaz, Iran
| | - Motahare Mokhtarzade
- Department of Sport Physiology, Shahid Chamran University, Ahvaz, Iran.,Department of Sport Physiology, Tarbiat Modares University, Tehran, Iran
| | | | - Andrej Fokin
- Institute of Sports Sciences and Innovation, Lithuanian Sports University, Kaunas, Lithuania
| |
Collapse
|
26
|
Abstract
There has been an ever-expanding list of the Limb-Girdle Muscular Dystrophies (LGMD). There are currently 8 subtypes of autosomal dominant (AD) and 26 subtypes of autosomal recessive (AR) LGMD. Despite continued research efforts to conquer this group of genetic neuromuscular disease, patients continue to be treated symptomatically with the aim of prevention or addressing complications. Mouse models have been helpful in clarifying disease pathogenesis as well as strategizing pathways for treatment. Discoveries in translational research as well as molecular therapeutic approaches have kept clinicians optimistic that more promising clinical trials will lead the way to finding the cure for these devastating disorders. It is well known that the challenge for these rare diseases is the ability to assemble adequate numbers of patients for a clinically meaningful trial, but current efforts in developing patient registries have been encouraging. Natural history studies will be essential in establishing and interpreting the appropriate outcome measures for clinical trials. Nevertheless, animal studies continue to be key in providing proof of concept that will be necessary in moving research along. This review will briefly discuss each type of LGMD, highlighting their distinguishing features, then focus on research efforts that have been published in the literature for the past few years, many of which are still in the preclinical trial stage.
Collapse
Affiliation(s)
- Mary Lynn Chu
- Department of Neurology, New York University School of Medicine, New York, New York, 10016, USA.
- New York University Langone Orthopedic Hospital, 301 East 17th Street, New York, New York, 10003, USA.
| | - Ellen Moran
- Division of Clinical Genetics, Center for Children, Hassenfeld Children's Hospital at New York University Langone, New York University Langone Orthopedic Hospital, 301 East 17th Street, New York, New York, 10003, USA
| |
Collapse
|
27
|
Kristina Parr M, Müller-Schöll A. Pharmacology of doping agents—mechanisms promoting muscle hypertrophy. AIMS MOLECULAR SCIENCE 2018. [DOI: 10.3934/molsci.2018.2.131] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
|
28
|
Past, Present, and Future Perspective of Targeting Myostatin and Related Signaling Pathways to Counteract Muscle Atrophy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1088:153-206. [DOI: 10.1007/978-981-13-1435-3_8] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
29
|
Barreto R, Waning DL, Gao H, Liu Y, Zimmers TA, Bonetto A. Chemotherapy-related cachexia is associated with mitochondrial depletion and the activation of ERK1/2 and p38 MAPKs. Oncotarget 2017; 7:43442-43460. [PMID: 27259276 PMCID: PMC5190036 DOI: 10.18632/oncotarget.9779] [Citation(s) in RCA: 144] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 05/16/2016] [Indexed: 12/21/2022] Open
Abstract
Cachexia affects the majority of cancer patients, with currently no effective treatments. Cachexia is defined by increased fatigue and loss of muscle function resulting from muscle and fat depletion. Previous studies suggest that chemotherapy may contribute to cachexia, although the causes responsible for this association are not clear. The purpose of this study was to investigate the mechanism(s) associated with chemotherapy-related effects on body composition and muscle function. Normal mice were administered chemotherapy regimens used for the treatment of colorectal cancer, such as Folfox (5-FU, leucovorin, oxaliplatin) or Folfiri (5-FU, leucovorin, irinotecan) for 5 weeks. The animals that received chemotherapy exhibited concurrent loss of muscle mass and muscle weakness. Consistently with previous findings, muscle wasting was associated with up-regulation of ERK1/2 and p38 MAPKs. No changes in ubiquitin-dependent proteolysis or in the expression of TGFβ-family members were detected. Further, marked decreases in mitochondrial content, associated with abnormalities at the sarcomeric level and with increase in the number of glycolytic fibers were observed in the muscle of mice receiving chemotherapy. Finally, ACVR2B/Fc or PD98059 prevented Folfiri-associated ERK1/2 activation and myofiber atrophy in C2C12 cultures. Our findings demonstrate that chemotherapy promotes MAPK-dependent muscle atrophy as well as mitochondrial depletion and alterations of the sarcomeric units. Therefore, these findings suggest that chemotherapy potentially plays a causative role in the occurrence of muscle loss and weakness. Moreover, the present observations provide a strong rationale for testing ACVR2B/Fc or MEK1 inhibitors in combination with anticancer drugs as novel strategies aimed at preventing chemotherapy-associated muscle atrophy.
Collapse
Affiliation(s)
- Rafael Barreto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - David L Waning
- Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,IUPUI Center for Cachexia Research, Innovation and Therapy, Indianapolis, IN 46202, USA
| | - Hongyu Gao
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yunlong Liu
- Department of Medical and Molecular Genetics, Center for Computational Biology and Bioinformatics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Teresa A Zimmers
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,IUPUI Center for Cachexia Research, Innovation and Therapy, Indianapolis, IN 46202, USA
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.,IUPUI Center for Cachexia Research, Innovation and Therapy, Indianapolis, IN 46202, USA
| |
Collapse
|
30
|
Barreto R, Kitase Y, Matsumoto T, Pin F, Colston KC, Couch KE, O'Connell TM, Couch ME, Bonewald LF, Bonetto A. ACVR2B/Fc counteracts chemotherapy-induced loss of muscle and bone mass. Sci Rep 2017; 7:14470. [PMID: 29089584 PMCID: PMC5665981 DOI: 10.1038/s41598-017-15040-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/20/2017] [Indexed: 01/06/2023] Open
Abstract
Chemotherapy promotes the development of cachexia, a debilitating condition characterized by muscle and fat loss. ACVR2B/Fc, an inhibitor of the Activin Receptor 2B signaling, has been shown to preserve muscle mass and prolong survival in tumor hosts, and to increase bone mass in models of osteogenesis imperfecta and muscular dystrophy. We compared the effects of ACVR2B/Fc on muscle and bone mass in mice exposed to Folfiri. In addition to impairing muscle mass and function, Folfiri had severe negative effects on bone, as shown by reduced trabecular bone volume fraction (BV/TV), thickness (Tb.Th), number (Tb.N), connectivity density (Conn.Dn), and by increased separation (Tb.Sp) in trabecular bone of the femur and vertebra. ACVR2B/Fc prevented the loss of muscle mass and strength, and the loss of trabecular bone in femurs and vertebrae following Folfiri administration. Neither Folfiri nor ACVR2B/Fc had effects on femoral cortical bone, as shown by unchanged cortical bone volume fraction (Ct.BV/TV), thickness (Ct.Th) and porosity. Our results suggest that Folfiri is responsible for concomitant muscle and bone degeneration, and that ACVR2B/Fc prevents these derangements. Future studies are required to determine if the same protective effects are observed in combination with other anticancer regimens or in the presence of cancer.
Collapse
Affiliation(s)
- Rafael Barreto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Yukiko Kitase
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Tsutomu Matsumoto
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Fabrizio Pin
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Kyra C Colston
- Indianapolis Project STEM, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Katherine E Couch
- Indianapolis Project STEM, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Thomas M O'Connell
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Otolaryngology - Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Marion E Couch
- Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Department of Otolaryngology - Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Lynda F Bonewald
- Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.,IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN, 46202, USA
| | - Andrea Bonetto
- Department of Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Anatomy and Cell Biology, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Indiana Center for Musculoskeletal Health, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Department of Otolaryngology - Head and Neck Surgery, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,Simon Cancer Center, Indiana University School of Medicine, Indianapolis, IN, 46202, USA. .,IUPUI Center for Cachexia Research Innovation and Therapy, Indiana University School of Medicine, Indianapolis, IN, 46202, USA.
| |
Collapse
|
31
|
Ma J, Pichavant C, du Bois H, Bhakta M, Calos MP. DNA-Mediated Gene Therapy in a Mouse Model of Limb Girdle Muscular Dystrophy 2B. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 7:123-131. [PMID: 29159199 PMCID: PMC5684445 DOI: 10.1016/j.omtm.2017.10.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 10/19/2017] [Indexed: 11/26/2022]
Abstract
Mutations in the gene for dysferlin cause a degenerative disorder of skeletal muscle known as limb girdle muscular dystrophy 2B. To achieve gene delivery of plasmids encoding dysferlin to hind limb muscles of dysferlin knockout mice, we used a vascular injection method that perfused naked plasmid DNA into all major muscle groups of the hind limb. We monitored delivery by luciferase live imaging and western blot, confirming strong dysferlin expression that persisted over the 3-month time course of the experiment. Co-delivery of the follistatin gene, which may promote muscle growth, was monitored by ELISA. Immunohistochemistry documented the presence of dysferlin in muscle fibers in treated limbs, and PCR confirmed the presence of plasmid DNA. Because dysferlin is involved in repair of the sarcolemmal membrane, dysferlin loss leads to fragile sarcolemmal membranes that can be detected by permeability to Evan’s blue dye. We showed that after gene therapy with a plasmid encoding both dysferlin and follistatin, statistically significant reduction in Evan’s blue dye permeability was present in hamstring muscles. These results suggest that vascular delivery of plasmids carrying these therapeutic genes may lead to simple and effective approaches for improving the clinical condition of limb girdle muscular dystrophy 2B.
Collapse
Affiliation(s)
- Julia Ma
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | - Christophe Pichavant
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | - Haley du Bois
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | - Mital Bhakta
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| | - Michele P Calos
- Department of Genetics, Stanford University School of Medicine, Stanford, CA 94305-5120, USA
| |
Collapse
|
32
|
Silencing Nfix rescues muscular dystrophy by delaying muscle regeneration. Nat Commun 2017; 8:1055. [PMID: 29057908 PMCID: PMC5651883 DOI: 10.1038/s41467-017-01098-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2016] [Accepted: 08/17/2017] [Indexed: 11/30/2022] Open
Abstract
Muscular dystrophies are severe disorders due to mutations in structural genes, and are characterized by skeletal muscle wasting, compromised patient mobility, and respiratory functions. Although previous works suggested enhancing regeneration and muscle mass as therapeutic strategies, these led to no long-term benefits in humans. Mice lacking the transcription factor Nfix have delayed regeneration and a shift toward an oxidative fiber type. Here, we show that ablating or silencing the transcription factor Nfix ameliorates pathology in several forms of muscular dystrophy. Silencing Nfix in postnatal dystrophic mice, when the first signs of the disease already occurred, rescues the pathology and, conversely, Nfix overexpression in dystrophic muscles increases regeneration and markedly exacerbates the pathology. We therefore offer a proof of principle for a novel therapeutic approach for muscular dystrophies based on delaying muscle regeneration. Strategies aimed at promoting muscle regeneration to treat muscular dystrophy have met with limited success. Here the authors show instead that delaying muscle regeneration, by ablation of the transcription factor Nfix, ameliorates muscular dystrophy in mice.
Collapse
|
33
|
Quattrocelli M, Salamone IM, Page PG, Warner JL, Demonbreun AR, McNally EM. Intermittent Glucocorticoid Dosing Improves Muscle Repair and Function in Mice with Limb-Girdle Muscular Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2017; 187:2520-2535. [PMID: 28823869 DOI: 10.1016/j.ajpath.2017.07.017] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 07/03/2017] [Accepted: 07/13/2017] [Indexed: 12/17/2022]
Abstract
The muscular dystrophies are genetically diverse. Shared pathological features among muscular dystrophies include breakdown, or loss of muscle, and accompanying fibrotic replacement. Novel strategies are needed to enhance muscle repair and function and to slow this pathological remodeling. Glucocorticoid steroids, like prednisone, are known to delay loss of ambulation in patients with Duchenne muscular dystrophy but are accompanied by prominent adverse effects. However, less is known about the effects of steroid administration in other types of muscular dystrophies, including limb-girdle muscular dystrophies (LGMDs). LGMD 2B is caused by loss of dysferlin, a membrane repair protein, and LGMD 2C is caused by loss of the dystrophin-associated protein, γ-sarcoglycan. Herein, we assessed the efficacy of steroid dosing on sarcolemmal repair, muscle function, histopathology, and the regenerative capacity of primary muscle cells. We found that in murine models of LGMD 2B and 2C, daily prednisone dosing reduced muscle damage and fibroinflammatory infiltration. However, daily prednisone dosing also correlated with increased muscle adipogenesis and atrophic remodeling. Conversely, intermittent dosing of prednisone, provided once weekly, enhanced muscle repair and did not induce atrophy or adipogenesis, and was associated with improved muscle function. These data indicate that dosing frequency of glucocorticoid steroids affects muscle remodeling in non-Duchenne muscular dystrophies, suggesting a positive outcome associated with intermittent steroid dosing in LGMD 2B and 2C muscle.
Collapse
Affiliation(s)
- Mattia Quattrocelli
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Isabella M Salamone
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Patrick G Page
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - James L Warner
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Alexis R Demonbreun
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Elizabeth M McNally
- Center for Genetic Medicine, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
34
|
Nissinen TA, Degerman J, Räsänen M, Poikonen AR, Koskinen S, Mervaala E, Pasternack A, Ritvos O, Kivelä R, Hulmi JJ. Systemic blockade of ACVR2B ligands prevents chemotherapy-induced muscle wasting by restoring muscle protein synthesis without affecting oxidative capacity or atrogenes. Sci Rep 2016; 6:32695. [PMID: 27666826 PMCID: PMC5036092 DOI: 10.1038/srep32695] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 08/12/2016] [Indexed: 12/26/2022] Open
Abstract
Doxorubicin is a widely used and effective chemotherapy drug. However, cardiac and skeletal muscle toxicity of doxorubicin limits its use. Inhibiting myostatin/activin signalling can prevent muscle atrophy, but its effects in chemotherapy-induced muscle wasting are unknown. In the present study we investigated the effects of doxorubicin administration alone or combined with activin receptor ligand pathway blockade by soluble activin receptor IIB (sACVR2B-Fc). Doxorubicin administration decreased body mass, muscle size and bone mineral density/content in mice. However, these effects were prevented by sACVR2B-Fc administration. Unlike in many other wasting situations, doxorubicin induced muscle atrophy without markedly increasing typical atrogenes or protein degradation pathways. Instead, doxorubicin decreased muscle protein synthesis which was completely restored by sACVR2B-Fc. Doxorubicin administration also resulted in impaired running performance without effects on skeletal muscle mitochondrial capacity/function or capillary density. Running performance and mitochondrial function were unaltered by sACVR2B-Fc administration. Tumour experiment using Lewis lung carcinoma cells demonstrated that sACVR2B-Fc decreased the cachectic effects of chemotherapy without affecting tumour growth. These results demonstrate that blocking ACVR2B signalling may be a promising strategy to counteract chemotherapy-induced muscle wasting without damage to skeletal muscle oxidative capacity or cancer treatment.
Collapse
Affiliation(s)
- T A Nissinen
- Department of Biology of Physical Activity, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - J Degerman
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - M Räsänen
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - A R Poikonen
- Department of Biology of Physical Activity, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland
| | - S Koskinen
- LIKES Research Center for Sport and Health Sciences, Jyväskylä, Finland
| | - E Mervaala
- Department of Pharmacology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - A Pasternack
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland
| | - O Ritvos
- Department of Bacteriology and Immunology, Haartman Institute, University of Helsinki, Helsinki, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - R Kivelä
- Wihuri Research Institute and Translational Cancer Biology Program, University of Helsinki, Helsinki, Finland
| | - J J Hulmi
- Department of Biology of Physical Activity, Neuromuscular Research Center, University of Jyväskylä, Jyväskylä, Finland.,Department of Physiology, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| |
Collapse
|
35
|
Cárdenas AM, González-Jamett AM, Cea LA, Bevilacqua JA, Caviedes P. Dysferlin function in skeletal muscle: Possible pathological mechanisms and therapeutical targets in dysferlinopathies. Exp Neurol 2016; 283:246-54. [PMID: 27349407 DOI: 10.1016/j.expneurol.2016.06.026] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Revised: 06/22/2016] [Accepted: 06/23/2016] [Indexed: 12/18/2022]
Abstract
Mutations in the dysferlin gene are linked to a group of muscular dystrophies known as dysferlinopathies. These myopathies are characterized by progressive atrophy. Studies in muscle tissue from dysferlinopathy patients or dysferlin-deficient mice point out its importance in membrane repair. However, expression of dysferlin homologous proteins that restore sarcolemma repair function in dysferlinopathy animal models fail to arrest muscle wasting, therefore suggesting that dysferlin plays other critical roles in muscle function. In the present review, we discuss dysferlin functions in the skeletal muscle, as well as pathological mechanisms related to dysferlin mutations. Particular focus is presented related the effect of dysferlin on cell membrane related function, which affect its repair, vesicle trafficking, as well as Ca(2+) homeostasis. Such mechanisms could provide accessible targets for pharmacological therapies.
Collapse
Affiliation(s)
- Ana M Cárdenas
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile.
| | - Arlek M González-Jamett
- Centro Interdisciplinario de Neurociencia de Valparaíso, Facultad de Ciencias, Universidad de Valparaíso, Valparaíso, Chile; Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Luis A Cea
- Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Jorge A Bevilacqua
- Programa de Anatomía y Biología del Desarrollo, ICBM, Facultad de Medicina, Departamento de Neurología y Neurocirugía, Hospital Clínico Universidad de Chile, Universidad de Chile, Santiago, Chile
| | - Pablo Caviedes
- Programa de Farmacología Molecular y Clinica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| |
Collapse
|
36
|
Kornegay JN, Bogan DJ, Bogan JR, Dow JL, Wang J, Fan Z, Liu N, Warsing LC, Grange RW, Ahn M, Balog-Alvarez CJ, Cotten SW, Willis MS, Brinkmeyer-Langford C, Zhu H, Palandra J, Morris CA, Styner MA, Wagner KR. Dystrophin-deficient dogs with reduced myostatin have unequal muscle growth and greater joint contractures. Skelet Muscle 2016; 6:14. [PMID: 27047655 PMCID: PMC4819282 DOI: 10.1186/s13395-016-0085-7] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 02/24/2016] [Indexed: 09/02/2023] Open
Abstract
Background Myostatin (Mstn) is a negative regulator of muscle growth whose inhibition promotes muscle growth and regeneration. Dystrophin-deficient mdx mice in which myostatin is knocked out or inhibited postnatally have a less severe phenotype with greater total mass and strength and less fibrosis and fatty replacement of muscles than mdx mice with wild-type myostatin expression. Dogs with golden retriever muscular dystrophy (GRMD) have previously been noted to have increased muscle mass and reduced fibrosis after systemic postnatal myostatin inhibition. Based partly on these results, myostatin inhibitors are in development for use in human muscular dystrophies. However, persisting concerns regarding the effects of long-term and profound myostatin inhibition will not be easily or imminently answered in clinical trials. Methods To address these concerns, we developed a canine (GRippet) model by crossbreeding dystrophin-deficient GRMD dogs with Mstn-heterozygous (Mstn+/−) whippets. A total of four GRippets (dystrophic and Mstn+/−), three GRMD (dystrophic and Mstn wild-type) dogs, and three non-dystrophic controls from two litters were evaluated. Results Myostatin messenger ribonucleic acid (mRNA) and protein levels were downregulated in both GRMD and GRippet dogs. GRippets had more severe postural changes and larger (more restricted) maximal joint flexion angles, apparently due to further exaggeration of disproportionate effects on muscle size. Flexors such as the cranial sartorius were more hypertrophied on magnetic resonance imaging (MRI) in the GRippets, while extensors, including the quadriceps femoris, underwent greater atrophy. Myostatin protein levels negatively correlated with relative cranial sartorius muscle cross-sectional area on MRI, supporting a role in disproportionate muscle size. Activin receptor type IIB (ActRIIB) expression was higher in dystrophic versus control dogs, consistent with physiologic feedback between myostatin and ActRIIB. However, there was no differential expression between GRMD and GRippet dogs. Satellite cell exhaustion was not observed in GRippets up to 3 years of age. Conclusions Partial myostatin loss may exaggerate selective muscle hypertrophy or atrophy/hypoplasia in GRMD dogs and worsen contractures. While muscle imbalance is not a feature of myostatin inhibition in mdx mice, findings in a larger animal model could translate to human experience with myostatin inhibitors. Electronic supplementary material The online version of this article (doi:10.1186/s13395-016-0085-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Joe N Kornegay
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ; Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ; Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458 USA
| | - Daniel J Bogan
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Janet R Bogan
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Jennifer L Dow
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Jiahui Wang
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Zheng Fan
- Department of Neurology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Naili Liu
- The Hugo W. Moser Research Institute at Kennedy Krieger Institute and Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Leigh C Warsing
- The Hugo W. Moser Research Institute at Kennedy Krieger Institute and Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205 USA
| | - Robert W Grange
- Department of Human Nutrition, Foods and Exercise, Virginia Tech University, Blacksburg, VA 24061 USA
| | - Mihye Ahn
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Cynthia J Balog-Alvarez
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458 USA
| | - Steven W Cotten
- Department of Pathology, The Ohio State University, Columbus, OH 43210 USA
| | - Monte S Willis
- Department of Pathology and Laboratory Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Candice Brinkmeyer-Langford
- Department of Veterinary Integrative Biosciences, College of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX 77843-4458 USA
| | - Hongtu Zhu
- Department of Biostatistics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Joe Palandra
- Rare Disease Research Unit, Pfizer, Inc., Cambridge Park Drive, Cambridge, MA USA
| | - Carl A Morris
- Rare Disease Research Unit, Pfizer, Inc., Cambridge Park Drive, Cambridge, MA USA
| | - Martin A Styner
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA ; Department of Computer Science, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599 USA
| | - Kathryn R Wagner
- The Hugo W. Moser Research Institute at Kennedy Krieger Institute and Departments of Neurology and Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
37
|
Genome-wide expression analysis comparing hypertrophic changes in normal and dysferlinopathy mice. GENOMICS DATA 2015; 6:253-7. [PMID: 26697388 PMCID: PMC4664771 DOI: 10.1016/j.gdata.2015.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Accepted: 10/18/2015] [Indexed: 11/25/2022]
Abstract
Because myostatin normally limits skeletal muscle growth, there are extensive efforts to develop myostatin inhibitors for clinical use. One potential concern is that in muscle degenerative diseases, inducing hypertrophy may increase stress on dystrophic fibers. Our study shows that blocking this pathway in dysferlin deficient mice results in early improvement in histopathology but ultimately accelerates muscle degeneration. Hence, benefits of this approach should be weighed against these potential detrimental effects. Here, we present detailed experimental methods and analysis for the gene expression profiling described in our recently published study in Human Molecular Genetics (Lee et al., 2015). Our data sets have been deposited in the Gene Expression Omnibus (GEO) database (GSE62945) and are available at http://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE62945. Our data provide a resource for exploring molecular mechanisms that are related to hypertrophy-induced, accelerated muscular degeneration in dysferlinopathy.
Collapse
|
38
|
Yue Y, Binalsheikh IM, Leach SB, Domeier TL, Duan D. Prospect of gene therapy for cardiomyopathy in hereditary muscular dystrophy. Expert Opin Orphan Drugs 2015; 4:169-183. [PMID: 27340611 DOI: 10.1517/21678707.2016.1124039] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Cardiac involvement is a common feature in muscular dystrophies. It presents as heart failure and/or arrhythmia. Traditionally, dystrophic cardiomyopathy is treated with symptom-relieving medications. Identification of disease-causing genes and investigation on pathogenic mechanisms have opened new opportunities to treat dystrophic cardiomyopathy with gene therapy. Replacing/repairing the mutated gene and/or targeting the pathogenic process/mechanisms using alternative genes may attenuate heart disease in muscular dystrophies. AREAS COVERED Duchenne muscular dystrophy is the most common muscular dystrophy. Duchenne cardiomyopathy has been the primary focus of ongoing dystrophic cardiomyopathy gene therapy studies. Here, we use Duchenne cardiomyopathy gene therapy to showcase recent developments and to outline the path forward. We also discuss gene therapy status for cardiomyopathy associated with limb-girdle and congenital muscular dystrophies, and myotonic dystrophy. EXPERT OPINION Gene therapy for dystrophic cardiomyopathy has taken a slow but steady path forward. Preclinical studies over the last decades have addressed many fundamental questions. Adeno-associated virus-mediated gene therapy has significantly improved the outcomes in rodent models of Duchenne and limb girdle muscular dystrophies. Validation of these encouraging results in large animal models will pave the way to future human trials.
Collapse
Affiliation(s)
- Yongping Yue
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri
| | | | - Stacey B Leach
- Department of Veterinary Medicine and Surgery, College of Veterinary Medicine, University of Missouri
| | - Timothy L Domeier
- Department of Medical Physiology and Pharmacology, School of Medicine, University of Missouri
| | - Dongsheng Duan
- Department of Molecular Microbiology and Immunology, School of Medicine, University of Missouri; Department of Neurology, School of Medicine, University of Missouri
| |
Collapse
|