1
|
Henzi BC, Lava SAG, Spagnuolo C, Putananickal N, Donner BC, Pfluger M, Burkhardt B, Fischer D. Tamoxifen may contribute to preserve cardiac function in Duchenne muscular dystrophy. Eur J Pediatr 2024; 183:4057-4062. [PMID: 38960907 PMCID: PMC11322393 DOI: 10.1007/s00431-024-05670-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/20/2024] [Accepted: 06/24/2024] [Indexed: 07/05/2024]
Abstract
Duchenne muscular dystrophy is life-limiting. Cardiomyopathy, which mostly ensues in the second decade of life, is the main cause of death. Treatment options are still limited. The TAMDMD (NCT03354039) trial assessed motor function, muscle strength and structure, laboratory biomarkers, and safety in 79 ambulant boys with genetically confirmed Duchenne muscular dystrophy, 6.5-12 years of age, receiving either daily tamoxifen 20 mg or placebo for 48 weeks. In this post-hoc analysis, available echocardiographic data of ambulant patients recruited at one study centre were retrieved and compared before and after treatment. Data from 14 patients, median 11 (interquartile range, IQR, 11-12) years of age was available. Baseline demographic characteristics were similar in participants assigned to placebo (n = 7) or tamoxifen (n = 7). Left ventricular end-diastolic diameter in the placebo group (median and IQR) was 39 (38-41) mm at baseline and 43 (38-44) mm at study end, while it was 44 (41-46) mm at baseline and 41 (37-46) mm after treatment in the tamoxifen group. Left ventricular fractional shortening in the placebo group was 35% (32-38%) before and 33% (32-36%) after treatment, while in the tamoxifen group it was 34% (33-34%) at baseline and 35% (33-35%) at study end. No safety signals were detected. CONCLUSION This hypothesis-generating post-hoc analysis suggests that tamoxifen over 48 weeks is well tolerated and may help preserving cardiac structure and function in Duchenne muscular dystrophy. Further studies are justified. CLINICALTRIALS gov Identifier: EudraCT 2017-004554-42, NCT03354039 What is known: • Duchenne muscular dystrophy (DMD) is life-limiting. Cardiomyopathy ensues in the second decade of life and is the main cause of death. Treatment options are still limited. • Tamoxifen reduced cardiac fibrosis in mice and improved cardiomyocyte function in human-induced pluripotent stem cell-derived cardiomyocytes. WHAT IS NEW • In this post-hoc analysis of the TAMDMD trial among 14 boys, median 11 years of age, treated with either tamoxifen or placebo for 48 weeks, treatment was well-tolerated. • A visual trend of improved left-ventricular dimensions and better systolic function preservation generates the hypothesis of a potential beneficial effect of tamoxifen in DMD cardiomyopathy.
Collapse
Affiliation(s)
- Bettina C Henzi
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
- Division of Neuropediatrics, Development and Rehabilitation, Department of Pediatrics, Inselspital, Bern University Hospital, and University of Bern, Bern, Switzerland
| | - Sebastiano A G Lava
- Pediatric Cardiology Unit, Department of Pediatrics, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Rue du Bugnon 46, Lausanne, 1011, Switzerland.
- Heart Failure and Transplantation, Department of Paediatric Cardiology, Great Ormond Street Hospital, London, UK.
- Clinical Pharmacology and Therapeutics Group, University College London, London, UK.
- Division of Clinical Pharmacology and Toxicology, Institute of Pharmacological Sciences of Southern Switzerland, Ente Ospedaliero Cantonale, Lugano, Switzerland.
| | - Carlos Spagnuolo
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Niveditha Putananickal
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| | - Birgit C Donner
- Pediatric Cardiology, University Children's Hospital Basel (UKBB), University of Basel, Basel, Switzerland
| | - Marc Pfluger
- Department of Cardiology, Center for Congenital Heart Disease, University Hospital of Bern, Bern, Switzerland
| | - Barbara Burkhardt
- Department of Pediatric Cardiology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Dirk Fischer
- Division of Neuropediatrics and Developmental Medicine, University Children's Hospital Basel, University of Basel, Basel, Switzerland
| |
Collapse
|
2
|
Gandhi S, Sweeney HL, Hart CC, Han R, Perry CGR. Cardiomyopathy in Duchenne Muscular Dystrophy and the Potential for Mitochondrial Therapeutics to Improve Treatment Response. Cells 2024; 13:1168. [PMID: 39056750 PMCID: PMC11274633 DOI: 10.3390/cells13141168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 07/05/2024] [Accepted: 07/06/2024] [Indexed: 07/28/2024] Open
Abstract
Duchenne muscular dystrophy (DMD) is a progressive neuromuscular disease caused by mutations to the dystrophin gene, resulting in deficiency of dystrophin protein, loss of myofiber integrity in skeletal and cardiac muscle, and eventual cell death and replacement with fibrotic tissue. Pathologic cardiac manifestations occur in nearly every DMD patient, with the development of cardiomyopathy-the leading cause of death-inevitable by adulthood. As early cardiac abnormalities are difficult to detect, timely diagnosis and appropriate treatment modalities remain a challenge. There is no cure for DMD; treatment is aimed at delaying disease progression and alleviating symptoms. A comprehensive understanding of the pathophysiological mechanisms is crucial to the development of targeted treatments. While established hypotheses of underlying mechanisms include sarcolemmal weakening, upregulation of pro-inflammatory cytokines, and perturbed ion homeostasis, mitochondrial dysfunction is thought to be a potential key contributor. Several experimental compounds targeting the skeletal muscle pathology of DMD are in development, but the effects of such agents on cardiac function remain unclear. The synergistic integration of small molecule- and gene-target-based drugs with metabolic-, immune-, or ion balance-enhancing compounds into a combinatorial therapy offers potential for treating dystrophin deficiency-induced cardiomyopathy, making it crucial to understand the underlying mechanisms driving the disorder.
Collapse
Affiliation(s)
- Shivam Gandhi
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
| | - H. Lee Sweeney
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA; (H.L.S.); (C.C.H.)
- Myology Institute, University of Florida, Gainesville, FL 32610, USA
| | - Cora C. Hart
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA; (H.L.S.); (C.C.H.)
- Myology Institute, University of Florida, Gainesville, FL 32610, USA
| | - Renzhi Han
- Department of Pediatrics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Christopher G. R. Perry
- School of Kinesiology and Health Science, Muscle Health Research Centre, York University, Toronto, ON M3J 1P3, Canada
| |
Collapse
|
3
|
Veeger TTJ, van de Velde NM, Keene KR, Niks EH, Hooijmans MT, Webb AG, de Groot JH, Kan HE. Baseline fat fraction is a strong predictor of disease progression in Becker muscular dystrophy. NMR IN BIOMEDICINE 2022; 35:e4691. [PMID: 35032073 PMCID: PMC9286612 DOI: 10.1002/nbm.4691] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/24/2021] [Accepted: 01/10/2022] [Indexed: 06/14/2023]
Abstract
In Becker muscular dystrophy (BMD), muscle weakness progresses relatively slowly, with a highly variable rate among patients. This complicates clinical trials, as clinically relevant changes are difficult to capture within the typical duration of a trial. Therefore, predictors for disease progression are needed. We assessed if temporal increase of fat fraction (FF) in BMD follows a sigmoidal trajectory and whether fat fraction at baseline (FFbase) could therefore predict FF increase after 2 years (ΔFF). Thereafter, for two different MR-based parameters, we tested the additional predictive value to FFbase. We used 3-T Dixon data from the upper and lower leg, and multiecho spin-echo MRI and 7-T 31 P MRS datasets from the lower leg, acquired in 24 BMD patients (age: 41.4 [SD 12.8] years). We assessed the pattern of increase in FF using mixed-effects modelling. Subsequently, we tested if indicators of muscle damage like standard deviation in water T2 (stdT2 ) and the phosphodiester (PDE) over ATP ratio at baseline had additional value to FFbase for predicting ∆FF. The association between FFbase and ΔFF was described by the derivative of a sigmoid function and resulted in a peak ΔFF around 0.45 FFbase (fourth-order polynomial term: t = 3.7, p < .001). StdT2 and PDE/ATP were not significantly associated with ∆FF if FFbase was included in the model. The relationship between FFbase and ∆FF suggests a sigmoidal trajectory of the increase in FF over time in BMD, similar to that described for Duchenne muscular dystrophy. Our results can be used to identify muscles (or patients) that are in the fast progressing stage of the disease, thereby facilitating the conduct of clinical trials.
Collapse
Affiliation(s)
- Thom T. J. Veeger
- C. J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Nienke M. van de Velde
- Department of Neurology, Leiden University Medical Center (LUMC)LeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| | - Kevin R. Keene
- Department of Neurology, Leiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Erik H. Niks
- Department of Neurology, Leiden University Medical Center (LUMC)LeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| | - Melissa T. Hooijmans
- Department of Radiology & Nuclear MedicineAmsterdam University Medical CentersAmsterdamThe Netherlands
| | - Andrew G. Webb
- C. J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Jurriaan H. de Groot
- Department of Rehabilitation Medicine, Leiden University Medical Center (LUMC)LeidenThe Netherlands
| | - Hermien E. Kan
- C. J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical Center (LUMC)LeidenThe Netherlands
- Duchenne Center NetherlandsThe Netherlands
| |
Collapse
|
4
|
Ma N, Chen D, Lee JH, Kuri P, Hernandez EB, Kocan J, Mahmood H, Tichy ED, Rompolas P, Mourkioti F. Piezo1 regulates the regenerative capacity of skeletal muscles via orchestration of stem cell morphological states. SCIENCE ADVANCES 2022; 8:eabn0485. [PMID: 35302846 PMCID: PMC8932657 DOI: 10.1126/sciadv.abn0485] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Accepted: 01/26/2022] [Indexed: 05/08/2023]
Abstract
Muscle stem cells (MuSCs) are essential for tissue homeostasis and regeneration, but the potential contribution of MuSC morphology to in vivo function remains unknown. Here, we demonstrate that quiescent MuSCs are morphologically heterogeneous and exhibit different patterns of cellular protrusions. We classified quiescent MuSCs into three functionally distinct stem cell states: responsive, intermediate, and sensory. We demonstrate that the shift between different stem cell states promotes regeneration and is regulated by the sensing protein Piezo1. Pharmacological activation of Piezo1 is sufficient to prime MuSCs toward more responsive cells. Piezo1 deletion in MuSCs shifts the distribution toward less responsive cells, mimicking the disease phenotype we find in dystrophic muscles. We further demonstrate that Piezo1 reactivation ameliorates the MuSC morphological and regenerative defects of dystrophic muscles. These findings advance our fundamental understanding of how stem cells respond to injury and identify Piezo1 as a key regulator for adjusting stem cell states essential for regeneration.
Collapse
Affiliation(s)
- Nuoying Ma
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Bioengineering Graduate Program, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Delia Chen
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ji-Hyung Lee
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paola Kuri
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Edward Blake Hernandez
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jacob Kocan
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hamd Mahmood
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elisia D. Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Panteleimon Rompolas
- Department of Dermatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Penn Institute for Regenerative Medicine, Musculoskeletal Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
5
|
Smith SJ, Fabian L, Sheikh A, Noche R, Cui X, Moore SA, Dowling JJ. Lysosomes and the pathogenesis of merosin-deficient congenital muscular dystrophy. Hum Mol Genet 2022; 31:733-747. [PMID: 34568901 PMCID: PMC9989739 DOI: 10.1093/hmg/ddab278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 09/01/2021] [Accepted: 09/14/2021] [Indexed: 11/14/2022] Open
Abstract
Congenital muscular dystrophy type 1A (MDC1A), the most common congenital muscular dystrophy in Western countries, is caused by recessive mutations in LAMA2, the gene encoding laminin alpha 2. Currently, no cure or disease modifying therapy has been successfully developed for MDC1A. Examination of patient muscle biopsies revealed altered distribution of lysosomes. We hypothesized that this redistribution was a novel and potentially druggable aspect of disease pathogenesis. We explored this hypothesis using candyfloss (caf), a zebrafish model of MDC1A. We found that lysosome distribution in caf zebrafish was also abnormal. This altered localization was significantly associated with fiber detachment and could be prevented by blocking myofiber detachment. Overexpression of transcription factor EB, a transcription factor that promotes lysosomal biogenesis, led to increased lysosome content and decreased fiber detachment. We conclude that genetic manipulation of the lysosomal compartment is able to alter the caf zebrafish disease process, suggesting that lysosome function may be a target for disease modification.
Collapse
Affiliation(s)
- Sarah J Smith
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Department of Family Medicine, University of Calgary, Calgary T2R 0X7, Alberta
| | - Lacramioara Fabian
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Adeel Sheikh
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Schulich School of Medicine and Dentistry, Western University, London, ON N6A 5C1, Canada
| | - Ramil Noche
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Zebrafish Genetics and Disease Models Core Facility, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Xiucheng Cui
- Zebrafish Genetics and Disease Models Core Facility, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
| | - Steven A Moore
- Department of Pathology, University of Iowa Medical Center, Iowa City, IA, USA
| | - James J Dowling
- Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 1A8, Canada
- Program for Genetics & Genome Biology, Hospital for Sick Children, Toronto, ON M5G 0A4, Canada
- Division of Neurology, Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
- Department of Paediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada
| |
Collapse
|
6
|
A fast Myosin super enhancer dictates muscle fiber phenotype through competitive interactions with Myosin genes. Nat Commun 2022; 13:1039. [PMID: 35210422 PMCID: PMC8873246 DOI: 10.1038/s41467-022-28666-1] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 02/04/2022] [Indexed: 12/15/2022] Open
Abstract
The contractile properties of adult myofibers are shaped by their Myosin heavy chain isoform content. Here, we identify by snATAC-seq a 42 kb super-enhancer at the locus regrouping the fast Myosin genes. By 4C-seq we show that active fast Myosin promoters interact with this super-enhancer by DNA looping, leading to the activation of a single promoter per nucleus. A rainbow mouse transgenic model of the locus including the super-enhancer recapitulates the endogenous spatio-temporal expression of adult fast Myosin genes. In situ deletion of the super-enhancer by CRISPR/Cas9 editing demonstrates its major role in the control of associated fast Myosin genes, and deletion of two fast Myosin genes at the locus reveals an active competition of the promoters for the shared super-enhancer. Last, by disrupting the organization of fast Myosin, we uncover positional heterogeneity within limb skeletal muscles that may underlie selective muscle susceptibility to damage in certain myopathies. The contractile properties of adult myofibers are shaped by their Myosin heavy chain isoform content. Here the authors show that a super enhancer controls the spatiotemporal expression of the genes at the fast myosin heavy chain locus by DNA looping and that this expression profile is recapitulated in a rainbow transgenic mouse model of the locus.
Collapse
|
7
|
Veeger TTJ, van Zwet EW, al Mohamad D, Naarding KJ, van de Velde NM, Hooijmans MT, Webb AG, Niks EH, de Groot JH, Kan HE. Muscle architecture is associated with muscle fat replacement in Duchenne and Becker muscular dystrophies. Muscle Nerve 2021; 64:576-584. [PMID: 34383334 PMCID: PMC9290788 DOI: 10.1002/mus.27399] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Revised: 08/02/2021] [Accepted: 08/07/2021] [Indexed: 12/21/2022]
Abstract
INTRODUCTION/AIMS Duchenne and Becker muscular dystrophies (DMD and BMD, respectively) are characterized by fat replacement of different skeletal muscles in a specific temporal order. Given the structural role of dystrophin in skeletal muscle mechanics, muscle architecture could be important in the progressive pathophysiology of muscle degeneration. Therefore, the aim of this study was to assess the role of muscle architecture in the progression of fat replacement in DMD and BMD. METHODS We assessed the association between literature-based leg muscle architectural characteristics and muscle fat fraction from 22 DMD and 24 BMD patients. Dixon-based magnetic resonance imaging estimates of fat fractions at baseline and 12 (only DMD) and 24 months were related to fiber length and physiological cross-sectional area (PCSA) using age-controlled linear mixed modeling. RESULTS DMD and BMD muscles with long fibers and BMD muscles with large PCSAs were associated with increased fat fraction. The effect of fiber length was stronger in muscles with larger PCSA. DISCUSSION Muscle architecture may explain the pathophysiology of muscle degeneration in dystrophinopathies, in which proximal muscles with a larger mass (fiber length × PCSA) are more susceptible, confirming the clinical observation of a temporal proximal-to-distal progression. These results give more insight into the mechanical role in the pathophysiology of muscular dystrophies. Ultimately, this new information can be used to help support the selection of current and the development of future therapies.
Collapse
Affiliation(s)
- Thom T. J. Veeger
- C.J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Erik W. van Zwet
- Department of BiostatisticsLeiden University Medical CenterLeidenThe Netherlands
| | - Diaa al Mohamad
- Department of BiostatisticsLeiden University Medical CenterLeidenThe Netherlands
| | - Karin J. Naarding
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
| | | | - Melissa T. Hooijmans
- Department of Radiology & Nuclear MedicineAmsterdam University Medical Centers, Location AMCAmsterdamThe Netherlands
| | - Andrew G. Webb
- C.J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| | - Erik H. Niks
- Department of NeurologyLeiden University Medical CenterLeidenThe Netherlands
| | - Jurriaan H. de Groot
- Department of Rehabilitation MedicineLeiden University Medical CenterLeidenThe Netherlands
| | - Hermien E. Kan
- C.J. Gorter Center for High Field MRI, Department of RadiologyLeiden University Medical CenterLeidenThe Netherlands
| |
Collapse
|
8
|
Tichy ED, Ma N, Sidibe D, Loro E, Kocan J, Chen DZ, Khurana TS, Hasty P, Mourkioti F. Persistent NF-κB activation in muscle stem cells induces proliferation-independent telomere shortening. Cell Rep 2021; 35:109098. [PMID: 33979621 PMCID: PMC8183356 DOI: 10.1016/j.celrep.2021.109098] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 11/30/2020] [Accepted: 04/16/2021] [Indexed: 12/30/2022] Open
Abstract
During the repeated cycles of damage and repair in many muscle disorders, including Duchenne muscular dystrophy (DMD), the muscle stem cell (MuSC) pool becomes less efficient at responding to and repairing damage. The underlying mechanism of such stem cell dysfunction is not fully known. Here, we demonstrate that the distinct early telomere shortening of diseased MuSCs in both mice and young DMD patients is associated with aberrant NF-κB activation. We find that prolonged NF-κB activation in MuSCs in chronic injuries leads to shortened telomeres and Ku80 dysregulation and results in severe skeletal muscle defects. Our studies provide evidence of a role for NF-κB in regulating stem-cell-specific telomere length, independently of cell replication, and could be a congruent mechanism that is applicable to additional tissues and/or diseases characterized by systemic chronic inflammation. Tichy et al. reveal a role for NF-κB signaling in regulating telomere length in muscle stem cells (MuSCs) after chronic injuries. Persistent activation of NF-κB leads to shortened telomeres, Ku80 dysregulation, and muscle defects. The findings link stem cell dysfunction and NF-κB-dependent telomere shortening in Duchenne muscular dystrophy.
Collapse
Affiliation(s)
- Elisia D Tichy
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nuoying Ma
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - David Sidibe
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Emanuele Loro
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jacob Kocan
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Delia Z Chen
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tejvir S Khurana
- Department of Physiology and Pennsylvania Muscle Institute, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Paul Hasty
- The Sam and Ann Barshop Institute for Longevity and Aging Studies, UT Health Science Center at San Antonio, San Antonio, TX 78229, USA
| | - Foteini Mourkioti
- Department of Orthopaedic Surgery, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA; Institute of Regenerative Medicine, Musculoskeletal Regeneration Program, Perelman School of Medicine, The University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
9
|
Nassoro DD, Torres L, Marando R, Mboma L, Mushi S, Habakkuk Mwakyula I. A child with duchenne muscular dystrophy: A case report of a rare diagnosis among Africans. Clin Case Rep 2020; 8:2654-2660. [PMID: 33363799 PMCID: PMC7752564 DOI: 10.1002/ccr3.3254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Revised: 07/20/2020] [Accepted: 07/22/2020] [Indexed: 01/14/2023] Open
Abstract
In Africa, lack of awareness and low index of suspicion of rare diseases like dystrophinopathies, directly or indirectly, contributes to the increased morbidity and mortality. Therefore, even though the data on prevalence is limited, we need to have a high degree of suspicion in patients presenting with suggestive clinical features.
Collapse
Affiliation(s)
- David D. Nassoro
- Department of Internal MedicineMbeya Zonal Referral HospitalMbeyaTanzania
- Department of Internal MedicineThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Liset Torres
- Department of PathologyMbeya Zonal Referral HospitalMbeyaTanzania
- Department of PathologyThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Rehema Marando
- Department of Pediatrics and Child HealthMbeya Zonal Referral HospitalMbeyaTanzania
- Department of Pediatrics and Child HealthThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Lazaro Mboma
- Department of SurgeryMbeya Zonal Referral HospitalMbeyaTanzania
- Department of SurgeryThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| | - Seraphine Mushi
- Department of PhysiotherapyMbeya Zonal Referral HospitalMbeyaTanzania
| | - Issakwisa Habakkuk Mwakyula
- Department of Internal MedicineMbeya Zonal Referral HospitalMbeyaTanzania
- Department of Internal MedicineThe University of Dar es SalaamMbeya College of Health and Allied SciencesMbeyaTanzania
| |
Collapse
|
10
|
Al-Khalili Szigyarto C. Duchenne Muscular Dystrophy: recent advances in protein biomarkers and the clinical application. Expert Rev Proteomics 2020; 17:365-375. [PMID: 32713262 DOI: 10.1080/14789450.2020.1773806] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Early biomarker discovery studies have praised the value of their emerging results, predicting an unprecedented impact on health care. Biomarkers are expected to provide tests with increased specificity and sensitivity compared to existing measures, improve the decision-making process, and accelerate the development of therapies. For rare disorders, like Duchenne Muscular Dystrophy (DMD) such biomarkers can assist the development of therapies, therefore also helping to find a cure for the disease. AREA COVERED State-of-the-art technologies have been used to identify blood biomarkers for DMD and efforts have been coordinated to develop and promote translation of biomarkers for clinical practice. Biomarker translation to clinical practice is however, adjoined by challenges related to the complexity of the disease, involving numerous biological processes, and the limited sample resources. This review highlights the current progress on the development of biomarkers, describing the proteomics technologies used, the most promising findings and the challenges encountered. EXPERT OPINION Strategies for effective use of samples combined with orthogonal proteomics methods for protein quantification are essential for translating biomarkers to the patient's bed side. Progress is achieved only if strong evidence is provided that the biomarker constitutes a reliable indicator of the patient's health status for a specific context of use.
Collapse
Affiliation(s)
- Cristina Al-Khalili Szigyarto
- Science for Life Laboratory, KTH - Royal Institute of Technology , Solna, Sweden.,School of Chemistry, Biotechnology and Health, KTH - Royal Institute of Technology , Stockholm, Sweden
| |
Collapse
|
11
|
Kim E, Wu F, Wu X, Choo HJ. Generation of craniofacial myogenic progenitor cells from human induced pluripotent stem cells for skeletal muscle tissue regeneration. Biomaterials 2020; 248:119995. [PMID: 32283390 PMCID: PMC7232788 DOI: 10.1016/j.biomaterials.2020.119995] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 03/13/2020] [Accepted: 03/20/2020] [Indexed: 12/18/2022]
Abstract
Craniofacial skeletal muscle is composed of approximately 60 muscles, which have critical functions including food uptake, eye movements and facial expressions. Although craniofacial muscles have significantly different embryonic origin, most current skeletal muscle differentiation protocols using human induced pluripotent stem cells (iPSCs) are based on somite-derived limb and trunk muscle developmental pathways. Since the lack of a protocol for craniofacial muscles is a significant gap in the iPSC-derived muscle field, we have developed an optimized protocol to generate craniofacial myogenic precursor cells (cMPCs) from human iPSCs by mimicking key signaling pathways during craniofacial embryonic myogenesis. At each different stage, human iPSC-derived cMPCs mirror the transcription factor expression profiles seen in their counterparts during embryo development. After the bi-potential cranial pharyngeal mesoderm is established, cells are committed to cranial skeletal muscle lineages with inhibition of cardiac lineages and are purified by flow cytometry. Furthermore, identities of Ipsc-derived cMPCs are verified with human primary myoblasts from craniofacial muscles using RNA sequencing. These data suggest that our new method could provide not only in vitro research tools to study muscle specificity of muscular dystrophy but also abundant and reliable cellular resources for tissue engineering to support craniofacial reconstruction surgery.
Collapse
Affiliation(s)
- Eunhye Kim
- Department of Cell Biology, School of Medcine, Emory University, Atlanta, GA, 30322, USA
| | - Fang Wu
- Department of Cell Biology, School of Medcine, Emory University, Atlanta, GA, 30322, USA
| | - Xuewen Wu
- Department of Otolaryngology-Head and Neck Surgery, School of Medicine, Emory University, Atlanta, GA, 30322, USA; Department of Otolaryngology Head and Neck Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Hyojung J Choo
- Department of Cell Biology, School of Medcine, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
12
|
van Putten M, Lloyd EM, de Greef JC, Raz V, Willmann R, Grounds MD. Mouse models for muscular dystrophies: an overview. Dis Model Mech 2020; 13:dmm043562. [PMID: 32224495 PMCID: PMC7044454 DOI: 10.1242/dmm.043562] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Muscular dystrophies (MDs) encompass a wide variety of inherited disorders that are characterized by loss of muscle tissue associated with a progressive reduction in muscle function. With a cure lacking for MDs, preclinical developments of therapeutic approaches depend on well-characterized animal models that recapitulate the specific pathology in patients. The mouse is the most widely and extensively used model for MDs, and it has played a key role in our understanding of the molecular mechanisms underlying MD pathogenesis. This has enabled the development of therapeutic strategies. Owing to advancements in genetic engineering, a wide variety of mouse models are available for the majority of MDs. Here, we summarize the characteristics of the most commonly used mouse models for a subset of highly studied MDs, collated into a table. Together with references to key publications describing these models, this brief but detailed overview would be useful for those interested in, or working with, mouse models of MD.
Collapse
Affiliation(s)
- Maaike van Putten
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | - Erin M Lloyd
- The University of Western Australia, School of Human Sciences, Perth 6009, Australia
| | - Jessica C de Greef
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | - Vered Raz
- Leiden University Medical Center, Department of Human Genetics, Leiden, 2333 ZA, The Netherlands
| | | | - Miranda D Grounds
- The University of Western Australia, School of Human Sciences, Perth 6009, Australia
| |
Collapse
|
13
|
Saclier M, Bonfanti C, Antonini S, Angelini G, Mura G, Zanaglio F, Taglietti V, Romanello V, Sandri M, Tonelli C, Petroni K, Cassano M, Messina G. Nutritional intervention with cyanidin hinders the progression of muscular dystrophy. Cell Death Dis 2020; 11:127. [PMID: 32071288 PMCID: PMC7028923 DOI: 10.1038/s41419-020-2332-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 02/03/2020] [Accepted: 02/04/2020] [Indexed: 12/25/2022]
Abstract
Muscular Dystrophies are severe genetic diseases due to mutations in structural genes, characterized by progressive muscle wasting that compromises patients' mobility and respiratory functions. Literature underlined oxidative stress and inflammation as key drivers of these pathologies. Interestingly among different myofiber classes, type I fibers display a milder dystrophic phenotype showing increased oxidative metabolism. This work shows the benefits of a cyanidin-enriched diet, that promotes muscle fiber-type switch and reduced inflammation in dystrophic alpha-sarcoglyan (Sgca) null mice having, as a net outcome, morphological and functional rescue. Notably, this benefit is achieved also when the diet is administered in dystrophic animals when the signs of the disease are seriously evident. Our work provides compelling evidence that a cyanidin-rich diet strongly delays the progression of muscular dystrophies, paving the way for a combinatorial approach where nutritional-based reduction of muscle inflammation and oxidative stress facilitate the successful perspectives of definitive treatments.
Collapse
Affiliation(s)
- Marielle Saclier
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Chiara Bonfanti
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Stefania Antonini
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Giuseppe Angelini
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Giada Mura
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Federica Zanaglio
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Valentina Taglietti
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Vanina Romanello
- Venetian Institute of Molecular Medicine (VIMM), Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Marco Sandri
- Venetian Institute of Molecular Medicine (VIMM), Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Chiara Tonelli
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Katia Petroni
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Marco Cassano
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy
| | - Graziella Messina
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
14
|
Dardas Z, Swedan S, Al-Sheikh Qassem A, Azab B. The impact of exome sequencing on the diagnostic yield of muscular dystrophies in consanguineous families. Eur J Med Genet 2020; 63:103845. [PMID: 31953240 DOI: 10.1016/j.ejmg.2020.103845] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 12/04/2019] [Accepted: 01/11/2020] [Indexed: 10/25/2022]
Abstract
Muscular dystrophies (MDs) are a heterogeneous group of inherited disorders that are characterized by progressive skeletal muscle weakness and dystrophic changes on muscle biopsy. The broad genetic and clinical heterogeneity of MDs make the accurate diagnosis difficult via conventional approaches. This study investigated 23 patients from eight unrelated consanguineous families with MDs. Previous clinical assessments did not accurately clarify the type of their MD and/or misdiagnose them with another disease. Exome sequencing (ES) is an efficient, time-saving, and cost-effective tool, enabling disease-causing variant (DCV) detection in affected individuals. We investigated the use of ES to diagnose MD and discover the underlying genetic etiology. We achieved a remarkable diagnostic success rate of 87.5% (7 out of 8 families) which is the highest rate reported thus far compared to previous studies. We identified two novel pathogenic variants in DYSF gene (c.4179delG, c.1149+3G > C). The latter variant impacts the splicing machinery of DYSF mRNA. Moreover, we further assessed the pathogenicity of four recurrent variants ((DYSF, c.4076T > C), (GMPPB, c.458C > T), (SGCA, c.739G > A) (TTN, c.7331G > A), designated their neurological impact and added new phenotypes in patients with these variants. To our knowledge, this is the first study applying an ES-based comprehensive molecular diagnosis to Jordanian cohort with MDs. Our findings confirmed that ES is a powerful approach for the diagnosis of MD patients. This efficient method of molecular diagnosis is crucial for guiding patient clinical care, genetic counseling, and most importantly, paving the way for gene therapy which is currently in clinical trials.
Collapse
Affiliation(s)
- Zain Dardas
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan; Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan.
| | - Samer Swedan
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | | | - Belal Azab
- Department of Pathology and Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan; Human and Molecular Genetics, Medical College of Virginia, Virginia Commonwealth University, Richmond, VA, USA.
| |
Collapse
|
15
|
Abstract
Muscle stem cells, or satellite cells, are required for skeletal muscle maintenance, growth, and repair. Following satellite cell activation, several factors drive asymmetric cell division to generate a stem cell and a proliferative progenitor that forms new muscle. The balance between symmetric self-renewal and asymmetric division significantly impacts the efficiency of regeneration. In this Review, we discuss the relationship of satellite cell heterogeneity and the establishment of polarity to asymmetric division, as well as how these processes are impacted in homeostasis, aging, and disease. We also highlight therapeutic opportunities for targeting satellite cell polarity and self-renewal to stimulate muscle regeneration.
Collapse
|
16
|
Piper M, Gronostajski R, Messina G. Nuclear Factor One X in Development and Disease. Trends Cell Biol 2018; 29:20-30. [PMID: 30287093 DOI: 10.1016/j.tcb.2018.09.003] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 09/03/2018] [Accepted: 09/06/2018] [Indexed: 02/06/2023]
Abstract
The past decade has seen incredible advances in the field of stem cell biology that have greatly improved our understanding of development and provided important insights into pathological processes. Transcription factors (TFs) play a central role in mediating stem cell proliferation, quiescence, and differentiation. One TF that contributes to these processes is Nuclear Factor One X (NFIX). Recently, NFIX activity has been shown to be essential in multiple organ systems and to have important translational impacts for human health. Here, we describe recent studies showing the contribution of NFIX to muscle development and muscular dystrophies, hematopoiesis, cancer, and neural stem cell biology, highlighting the importance of this knowledge in the development of therapeutic targets.
Collapse
Affiliation(s)
- Michael Piper
- School of Biomedical Sciences, The University of Queensland, Brisbane, Queensland 4072, Australia.
| | - Richard Gronostajski
- Department of Biochemistry, Genetics, Genomics & Bioinformatics Graduate Program, New York State Center of Excellence in Bioinformatics and Life Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Graziella Messina
- Department of Biosciences, University of Milan, via Celoria 26, 20133, Milan, Italy.
| |
Collapse
|
17
|
Gomes JP, Coatti GC, Valadares MC, Assoni AF, Pelatti MV, Secco M, Zatz M. Human Adipose-Derived CD146+ Stem Cells Increase Life Span of a Muscular Dystrophy Mouse Model More Efficiently than Mesenchymal Stromal Cells. DNA Cell Biol 2018; 37:798-804. [DOI: 10.1089/dna.2018.4158] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Affiliation(s)
- Juliana P. Gomes
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Giuliana C. Coatti
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Marcos C. Valadares
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Amanda F. Assoni
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mayra V. Pelatti
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mariane Secco
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| | - Mayana Zatz
- Human Genome and Stem-Cell Research Center, Institute of Biosciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
18
|
Jones KM, O’Grady G, Rodrigues MJ, Ranta A, Roxburgh RH, Love DR, Theadom A. Impacts for Children Living with Genetic Muscle Disorders and their Parents – Findings from a Population-Based Study. J Neuromuscul Dis 2018; 5:341-352. [DOI: 10.3233/jnd-170287] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Affiliation(s)
- Kelly M. Jones
- National Institute for Stroke and Applied Neurosciences, School of Public Health and Psychosocial Studies, Faculty of Health and Environmental Studies, Auckland University of Technology, Auckland, New Zealand
| | - Gina O’Grady
- Paediatric Neurology, Starship Children’s Health, Auckland, New Zealand
| | - Miriam J. Rodrigues
- Neurology Department, Auckland City Hospital, Auckland, New Zealand and the Muscular Dystrophy Association of New Zealand, Auckland, New Zealand
| | - Anna Ranta
- Departmentof Neurology, Wellington Regional Hospital, University of Otago, New Zealand
| | - Richard H. Roxburgh
- Neurology Department, Auckland City Hospital, Auckland, New Zealand and the Centre for Brain Research, University of Auckland, Auckland, New Zealand
| | - Donald R. Love
- Diagnostic Genetics, LabPLUS, Auckland City Hospital, Auckland, New Zealand
| | - Alice Theadom
- National Institute for Stroke and Applied Neurosciences, School of Public Health and Psychosocial Studies, Faculty of Health and Environmental Studies, Auckland University of Technology, Auckland, New Zealand
| | | |
Collapse
|
19
|
Llano-Diez M, Ortez CI, Gay JA, Álvarez-Cabado L, Jou C, Medina J, Nascimento A, Jimenez-Mallebrera C. Digital PCR quantification of miR-30c and miR-181a as serum biomarkers for Duchenne muscular dystrophy. Neuromuscul Disord 2017; 27:15-23. [DOI: 10.1016/j.nmd.2016.11.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 11/06/2016] [Accepted: 11/08/2016] [Indexed: 10/20/2022]
|
20
|
Pasteuning-Vuhman S, Boertje-van der Meulen JW, van Putten M, Overzier M, Ten Dijke P, Kiełbasa SM, Arindrarto W, Wolterbeek R, Lezhnina KV, Ozerov IV, Aliper AM, Hoogaars WM, Aartsma-Rus A, Loomans CJM. New function of the myostatin/activin type I receptor (ALK4) as a mediator of muscle atrophy and muscle regeneration. FASEB J 2016; 31:238-255. [PMID: 27733450 PMCID: PMC5161514 DOI: 10.1096/fj.201600675r] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Accepted: 09/22/2016] [Indexed: 12/13/2022]
Abstract
Skeletal muscle fibrosis and impaired muscle regeneration are major contributors to muscle wasting in Duchenne muscular dystrophy (DMD). Muscle growth is negatively regulated by myostatin (MSTN) and activins. Blockage of these pathways may improve muscle quality and function in DMD. Antisense oligonucleotides (AONs) were designed specifically to block the function of ALK4, a key receptor for the MSTN/activin pathway in skeletal muscle. AON-induced exon skipping resulted in specific Alk4 down-regulation, inhibition of MSTN activity, and increased myoblast differentiation in vitro. Unexpectedly, a marked decrease in muscle mass (10%) was found after Alk4 AON treatment in mdx mice. In line with in vitro results, muscle regeneration was stimulated, and muscle fiber size decreased markedly. Notably, when Alk4 was down-regulated in adult wild-type mice, muscle mass decreased even more. RNAseq analysis revealed dysregulated metabolic functions and signs of muscle atrophy. We conclude that ALK4 inhibition increases myogenesis but also regulates the tight balance of protein synthesis and degradation. Therefore, caution must be used when developing therapies that interfere with MSTN/activin pathways.—Pasteuning-Vuhman, S., Boertje-van der Meulen, J. W., van Putten, M., Overzier, M., ten Dijke, P., Kiełbasa, S. M., Arindrarto, W., Wolterbeek, R., Lezhnina, K. V., Ozerov, I. V., Aliper, A. M., Hoogaars, W. M., Aartsma-Rus, A., Loomans, C. J. M. New function of the myostatin/activin type I receptor (ALK4) as a mediator of muscle atrophy and muscle regeneration.
Collapse
Affiliation(s)
| | | | - Maaike van Putten
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Maurice Overzier
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| | - Peter Ten Dijke
- Department of Molecular and Cell Biology Leiden University Medical Center, Leiden, The Netherlands.,Cancer Genomics Center, Leiden University Medical Center, Leiden, The Netherlands
| | - Szymon M Kiełbasa
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Wibowo Arindrarto
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ron Wolterbeek
- Department of Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, The Netherlands
| | - Ksenia V Lezhnina
- InSilico Medicine, Incorporated, Emerging Technology Centers, Johns Hopkins University, Baltimore, Maryland, USA; and
| | - Ivan V Ozerov
- InSilico Medicine, Incorporated, Emerging Technology Centers, Johns Hopkins University, Baltimore, Maryland, USA; and
| | - Aleksandr M Aliper
- InSilico Medicine, Incorporated, Emerging Technology Centers, Johns Hopkins University, Baltimore, Maryland, USA; and
| | - Willem M Hoogaars
- Department of Human Movement Sciences, Faculty of Behavioral and Movement Sciences, Move Research Institute Amsterdam, Vrije Universiteit (VU) Amsterdam, Amsterdam, The Netherlands
| | - Annemieke Aartsma-Rus
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands;
| | - Cindy J M Loomans
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
21
|
Adaptive Immune Response Impairs the Efficacy of Autologous Transplantation of Engineered Stem Cells in Dystrophic Dogs. Mol Ther 2016; 24:1949-1964. [PMID: 27506452 DOI: 10.1038/mt.2016.163] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 08/03/2016] [Indexed: 12/27/2022] Open
Abstract
Duchenne muscular dystrophy is the most common genetic muscular dystrophy. It is caused by mutations in the dystrophin gene, leading to absence of muscular dystrophin and to progressive degeneration of skeletal muscle. We have demonstrated that the exon skipping method safely and efficiently brings to the expression of a functional dystrophin in dystrophic CD133+ cells injected scid/mdx mice. Golden Retriever muscular dystrophic (GRMD) dogs represent the best preclinical model of Duchenne muscular dystrophy, mimicking the human pathology in genotypic and phenotypic aspects. Here, we assess the capacity of intra-arterial delivered autologous engineered canine CD133+ cells of restoring dystrophin expression in Golden Retriever muscular dystrophy. This is the first demonstration of five-year follow up study, showing initial clinical amelioration followed by stabilization in mild and severe affected Golden Retriever muscular dystrophy dogs. The occurrence of T-cell response in three Golden Retriever muscular dystrophy dogs, consistent with a memory response boosted by the exon skipped-dystrophin protein, suggests an adaptive immune response against dystrophin.
Collapse
|
22
|
Boström K, Ahlström G, Sunvisson H. Being the Next of Kin of an Adult Person With Muscular Dystrophy. Clin Nurs Res 2016; 15:86-104; discussion 105-6. [PMID: 16638828 DOI: 10.1177/1054773805285706] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
A chronic disorder affects all members of the family in various ways. The aim of this study is to elucidate the next of kin's ( N= 36) experiences when an adult family member has muscular dystrophy. The relationships were partner (36%, n= 14), parent (18%, n= 7), child (21%, n= 8), sibling (15%, n= 6), and other relative (3%, n= 1). Latent content analysis is employed and involves an interpretation of the interviewtext. The results showthe meaning of being close to a person with muscular dystrophy through the themes that emerged: exposure of the family; the span between obligation and love; being vigilant, protective, and supportive; and striving for an ordinary life. This study reveals a need for healthcare staff to understand the next of kin's narrated meaning of changes when a family member has a progressive disease.
Collapse
Affiliation(s)
- Katrin Boström
- Swedish Institute for Disability Research, Orebro University, Center for Rehabilitation Research, Orebro County Council
| | | | | |
Collapse
|
23
|
Cea LA, Puebla C, Cisterna BA, Escamilla R, Vargas AA, Frank M, Martínez-Montero P, Prior C, Molano J, Esteban-Rodríguez I, Pascual I, Gallano P, Lorenzo G, Pian H, Barrio LC, Willecke K, Sáez JC. Fast skeletal myofibers of mdx mouse, model of Duchenne muscular dystrophy, express connexin hemichannels that lead to apoptosis. Cell Mol Life Sci 2016; 73:2583-99. [PMID: 26803842 PMCID: PMC11108387 DOI: 10.1007/s00018-016-2132-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Revised: 12/15/2015] [Accepted: 01/07/2016] [Indexed: 02/07/2023]
Abstract
Skeletal muscles of patients with Duchenne muscular dystrophy (DMD) show numerous alterations including inflammation, apoptosis, and necrosis of myofibers. However, the molecular mechanism that explains these changes remains largely unknown. Here, the involvement of hemichannels formed by connexins (Cx HCs) was evaluated in skeletal muscle of mdx mouse model of DMD. Fast myofibers of mdx mice were found to express three connexins (39, 43 and 45) and high sarcolemma permeability, which was absent in myofibers of mdx Cx43(fl/fl)Cx45(fl/fl):Myo-Cre mice (deficient in skeletal muscle Cx43/Cx45 expression). These myofibers did not show elevated basal intracellular free Ca(2+) levels, immunoreactivity to phosphorylated p65 (active NF-κB), eNOS and annexin V/active Caspase 3 (marker of apoptosis) but presented dystrophin immunoreactivity. Moreover, muscles of mdx Cx43(fl/fl)Cx45(fl/fl):Myo-Cre mice exhibited partial decrease of necrotic features (big cells and high creatine kinase levels). Accordingly, these muscles showed similar macrophage infiltration as control mdx muscles. Nonetheless, the hanging test performance of mdx Cx43(fl/fl)Cx45(fl/fl):Myo-Cre mice was significantly better than that of control mdx Cx43(fl/fl)Cx45(fl/fl) mice. All three Cxs found in skeletal muscles of mdx mice were also detected in fast myofibers of biopsy specimens from patients with muscular dystrophy. Thus, reduction of Cx expression and/or function of Cx HCs may be potential therapeutic approaches to abrogate myofiber apoptosis in DMD.
Collapse
Affiliation(s)
- Luis A Cea
- Present: Program of Anatomy and Developmental Biology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile.
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile.
| | - Carlos Puebla
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile
| | - Bruno A Cisterna
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile
| | - Rosalba Escamilla
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile
| | - Aníbal A Vargas
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile
| | - Marina Frank
- Division of Molecular Genetics, Life and Medical Sciences Institute, University of Bonn, 53115, Bonn, Germany
| | | | - Carmen Prior
- Unidad de Genética Molecular-INGEMM, Hospital Universitario La Paz-IdIPAZ, Madrid, Spain
| | - Jesús Molano
- Unidad de Genética Molecular-INGEMM, Hospital Universitario La Paz-IdIPAZ, Madrid, Spain
| | | | - Ignacio Pascual
- Servicio de Neuropediatría, Hospital Universitario La Paz-IdIPAZ, Madrid, Spain
| | - Pía Gallano
- Servicio de Genética, Hospital Santa Creu i Sant Pablo-CIBERER, Barcelona, Spain
| | - Gustavo Lorenzo
- Servicio de Pediatria, "Ramón y Cajal" Hospital-IRYCIS, Madrid, Spain
| | - Héctor Pian
- Servicio de Anatomía Patológica, "Ramón y Cajal" Hospital-IRYCIS, Madrid, Spain
| | - Luis C Barrio
- Unidad de Neurología Experimental, "Ramón y Cajal" Hospital-IRYCIS, Madrid, Spain
| | - Klaus Willecke
- Division of Molecular Genetics, Life and Medical Sciences Institute, University of Bonn, 53115, Bonn, Germany
| | - Juan C Sáez
- Departamento de Fisiología, Pontificia Universidad Católica de Chile, Av. Libertador Bernardo O'Higgins 340, Santiago, Chile.
- Centro Interdisciplinario de Neurociencias de Valparaíso, Valparaíso, Chile.
| |
Collapse
|
24
|
Sánchez-Chapul L, Ángel-Muñoz MD, Ruano-Calderón L, Luna-Angulo A, Coral-Vázquez R, Hernández-Hernández Ó, Magaña JJ, León-Hernández SR, Escobar-Cedillo RE, Vargas S. Dysferlin quantification in monocytes for rapid screening for dysferlinopathies. Muscle Nerve 2016; 54:1064-1071. [PMID: 27104310 DOI: 10.1002/mus.25156] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2016] [Indexed: 12/15/2022]
Abstract
INTRODUCTION In this study, we determined normal levels of dysferlin expression in CD14+ monocytes by flow cytometry (FC) as a screening tool for dysferlinopathies. METHODS Monocytes from 183 healthy individuals and 29 patients were immunolabeled, run on an FACScalibur flow cytometer, and analyzed by FlowJo software. RESULTS The relative quantity of dysferlin was expressed as mean fluorescence intensity (MFI). Performance of this diagnostic test was assessed by calculating likelihood ratios at different MFI cut-off points, which allowed definition of 4 disease classification groups in a simplified algorithm. CONCLUSION The MFI value may differentiate patients with dysferlinopathy from healthy individuals; it may be a useful marker for screening purposes. Muscle Nerve 54: 1064-1071, 2016.
Collapse
Affiliation(s)
- Laura Sánchez-Chapul
- División de Neurociencias, Instituto Nacional de Rehabilitación (INR), Calz. Mexico-Xochimilco 289, Col. Arenal de Guadalupe, Del. Tlalpan, 14389, Mexico City, Mexico
| | - Miguel Del Ángel-Muñoz
- División de Neurociencias, Instituto Nacional de Rehabilitación (INR), Calz. Mexico-Xochimilco 289, Col. Arenal de Guadalupe, Del. Tlalpan, 14389, Mexico City, Mexico
| | - Luis Ruano-Calderón
- Subdirección de Enseñanza y Capacitación, Investigación y Calidad en Salud, Secretaría de Salud del Estado de Durango (SSED), Durango City, Mexico
| | - Alexandra Luna-Angulo
- División de Neurociencias, Instituto Nacional de Rehabilitación (INR), Calz. Mexico-Xochimilco 289, Col. Arenal de Guadalupe, Del. Tlalpan, 14389, Mexico City, Mexico
| | - Ramón Coral-Vázquez
- Sección de Posgrado, Escuela Superior de Medicina, Instituto Politécnico Nacional (ESM-IPN), Mexico City, Mexico
| | | | - Jonathan J Magaña
- Laboratorio de Medicina Genómica, Servicio de Genética, (INR), Mexico City, Mexico
| | | | | | - Steven Vargas
- Laboratorio de Patología Experimental, Instituto Nacional de Neurología y Neurocirugía (INNN) "Manuel Velasco Suárez", Mexico City, Mexico
| |
Collapse
|
25
|
López-Bastida J, Oliva-Moreno J, Linertová R, Serrano-Aguilar P. Social/economic costs and health-related quality of life in patients with rare diseases in Europe. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2016; 17 Suppl 1:1-5. [PMID: 27023708 DOI: 10.1007/s10198-016-0780-7] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Accepted: 01/13/2016] [Indexed: 05/07/2023]
Affiliation(s)
- Julio López-Bastida
- University of Castilla-La Mancha, Talavera de la Reina, Toledo, Spain.
- Red de Investigación en Servicios Sanitarios en Enfermedades Crónicas (REDISSEC), Madrid, Spain.
| | - Juan Oliva-Moreno
- Red de Investigación en Servicios Sanitarios en Enfermedades Crónicas (REDISSEC), Madrid, Spain
- University of Castilla-La Mancha, Toledo, Spain
| | - Renata Linertová
- Red de Investigación en Servicios Sanitarios en Enfermedades Crónicas (REDISSEC), Madrid, Spain
- Fundación Canaria de Investigación Sanitaria (FUNCANIS), Las Palmas de Gran Canaria, Spain
| | - Pedro Serrano-Aguilar
- Red de Investigación en Servicios Sanitarios en Enfermedades Crónicas (REDISSEC), Madrid, Spain
- Evaluation and Planning Service at Canary Islands Health Service (SESCS), Santa Cruz de Tenerife, Spain
| |
Collapse
|
26
|
Cavazza M, Kodra Y, Armeni P, De Santis M, López-Bastida J, Linertová R, Oliva-Moreno J, Serrano-Aguilar P, Posada-de-la-Paz M, Taruscio D, Schieppati A, Iskrov G, Péntek M, von der Schulenburg JMG, Kanavos P, Chevreul K, Persson U, Fattore G. Social/economic costs and health-related quality of life in patients with Duchenne muscular dystrophy in Europe. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2016; 17 Suppl 1:19-29. [PMID: 27038625 DOI: 10.1007/s10198-016-0782-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2015] [Accepted: 01/13/2016] [Indexed: 05/02/2023]
Abstract
OBJECTIVE The aim of this study was to determine the economic burden from a societal perspective and the health-related quality of life (HRQOL) of patients with Duchenne muscular dystrophy (DMD) in Europe. METHODS We conducted a cross-sectional study of patients with DMD from Bulgaria, France, Germany, Hungary, Italy, Spain, Sweden, and the UK. Data on demographic characteristics, healthcare resource utilization, informal care, labor productivity losses, and HRQOL were collected from the questionnaires completed by patients or their caregivers. HRQOL was measured with the EuroQol 5-domain (EQ-5D) questionnaire. Costs have been estimated from a societal perspective adopting a bottom-up approach. RESULTS A total of 422 questionnaires were included in the study; 268 of which were collected from patients with DMD and 154 from caregivers. The average annual cost per person in 2012 ranged from €7657 in Hungary to €58,704 in France. Direct non-healthcare costs are the main component of whole costs and informal care is the main driver of non-healthcare costs. Costs are also shown to differ between children and adults. With regard to HRQOL of adult patients, the EQ-5D VAS score and EQ-5D index scores were 50.5 and 0.24, respectively. The corresponding EQ-5D VAS and EQ-5D index scores for caregivers were 74.7 and 0.71, respectively. CONCLUSIONS We have estimated the average annual cost per patient with DMD in eight European countries adopting a social perspective, and to our knowledge this is the first study with such a wide perspective. The results on costs show a considerable gap between Eastern and Western European countries. Non-healthcare costs range from 64 to 89 % of overall costs and informal care is to a great extent the main driver of this cost category. The HRQOL of people with DMD is much lower than that of the general population.
Collapse
Affiliation(s)
- Marianna Cavazza
- Centre for Research on Health and Social Care Management (CERGAS), Bocconi University, Via Roentgen 1, 20136, Milan, Italy.
| | - Yllka Kodra
- National Centre for Rare Diseases, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Patrizio Armeni
- Centre for Research on Health and Social Care Management (CERGAS), Bocconi University, Via Roentgen 1, 20136, Milan, Italy
| | - Marta De Santis
- National Centre for Rare Diseases, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Julio López-Bastida
- University of Castilla-La Mancha, Talavera de la Reina, Toledo, Spain
- Red de Investigación en Servicios Sanitarios en Enfermedades Crónicas (REDISSEC), Madrid, Spain
| | - Renata Linertová
- Red de Investigación en Servicios Sanitarios en Enfermedades Crónicas (REDISSEC), Madrid, Spain
- Fundación Canaria de Investigación Sanitaria (FUNCANIS), Las Palmas de Gran Canaria, Spain
| | - Juan Oliva-Moreno
- Red de Investigación en Servicios Sanitarios en Enfermedades Crónicas (REDISSEC), Madrid, Spain
- University of Castilla-La Mancha, Toledo, Spain
| | - Pedro Serrano-Aguilar
- Red de Investigación en Servicios Sanitarios en Enfermedades Crónicas (REDISSEC), Madrid, Spain
- Evaluation and Planning Service at Canary Islands Health Service, Santa Cruz de Tenerife, Spain
| | | | - Domenica Taruscio
- National Centre for Rare Diseases, Istituto Superiore di Sanità (ISS), Rome, Italy
| | - Arrigo Schieppati
- Centro di Ricerche Cliniche per le Malattie Rare "Aldo e Cele Daccò", Mario Negri Institute for Pharmacological Research, Ranica, Bergamo, Italy
| | - Georgi Iskrov
- Institute of Rare Diseases, Plovdiv, Bulgaria
- Department of Social Medicine and Public Health, Faculty of Public Health, Medical University of Plovdiv, Plovdiv, Bulgaria
| | - Márta Péntek
- Department of Health Economics, Corvinus University of Budapest, Budapest, Hungary
| | | | - Panos Kanavos
- Department of Social Policy and LSE Health, London School of Economics and Political Science, London, UK
| | - Karine Chevreul
- URC Eco Ile de France, AP-HP, Paris, France
- Université Paris Diderot, Sorbonne Paris Cité, ECEVE, UMRS 1123, Paris, France
- INSERM, ECEVE, U1123, Paris, France
| | - Ulf Persson
- The Swedish Institute for Health Economics, Lund, Sweden
| | - Giovanni Fattore
- Centre for Research on Health and Social Care Management (CERGAS), Bocconi University, Via Roentgen 1, 20136, Milan, Italy
| |
Collapse
|
27
|
Ruf-Zamojski F, Trivedi V, Fraser SE, Trinh LA. Spatio-Temporal Differences in Dystrophin Dynamics at mRNA and Protein Levels Revealed by a Novel FlipTrap Line. PLoS One 2015; 10:e0128944. [PMID: 26083378 PMCID: PMC4471274 DOI: 10.1371/journal.pone.0128944] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Accepted: 05/01/2015] [Indexed: 11/18/2022] Open
Abstract
Dystrophin (Dmd) is a structural protein that links the extracellular matrix to actin filaments in muscle fibers and is required for the maintenance of muscles integrity. Mutations in Dmd lead to muscular dystrophies in humans and other vertebrates. Here, we report the characterization of a zebrafish gene trap line that fluorescently labels the endogenous Dmd protein (Dmd-citrine, Gt(dmd-citrine) ct90a). We show that the Dmd-citrine line recapitulates endogenous dmd transcript expression and Dmd protein localization. Using this Dmd-citrine line, we follow Dmd localization to the myosepta in real-time using time-lapse microscopy, and find that the accumulation of Dmd protein at the transverse myosepta coincides with the onset of myotome formation, a critical stage in muscle maturation. We observed that Dmd protein localizes specifically to the myosepta prior to dmd mRNA localization. Additionally, we demonstrate that the Dmd-citrine line can be used to assess muscular dystrophy following both genetic and physical disruptions of the muscle.
Collapse
Affiliation(s)
- Frederique Ruf-Zamojski
- California Institute of Technology, Biological Imaging Center, Beckman Institute, Division of Biology, Pasadena, California 91125, United States of America
| | - Vikas Trivedi
- California Institute of Technology, Department of Bioengineering, Pasadena, California 91125, United States of America
| | - Scott E. Fraser
- California Institute of Technology, Biological Imaging Center, Beckman Institute, Division of Biology, Pasadena, California 91125, United States of America
| | - Le A. Trinh
- California Institute of Technology, Biological Imaging Center, Beckman Institute, Division of Biology, Pasadena, California 91125, United States of America
- * E-mail:
| |
Collapse
|
28
|
Abstract
Dystrophinopathies comprise a group of hereditary muscle disorders characterized by progressive wasting and weakness of skeletal muscle, as a result of degeneration of muscle fibers, and can be distinguished by the mode of transmission, age at onset and pattern of muscle weakness. The range of phenotypes associated with the region Xp21 has been expanding since identification of the gene in 1987. The mild end of the spectrum includes the phenotype of the muscle cramps with myoglobinuria and isolated quadriceps myopathy, while at the severe end, there are progressive muscle diseases that are classified as Duchenne / Becker muscular dystrophy (DMD/BMD).
Collapse
|
29
|
Aho AC, Hultsjö S, Hjelm K. Young adults' experiences of living with recessive limb-girdle muscular dystrophy from a salutogenic orientation: an interview study. Disabil Rehabil 2015; 37:2083-91. [PMID: 25582030 DOI: 10.3109/09638288.2014.998782] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE To describe young adults' experiences of living with recessive limb-girdle muscular dystrophy (LGMD2) from a salutogenic orientation. METHODS A qualitative explorative interview study, including 14 participants aged 20-30 years, was performed focusing on comprehensibility, manageability and meaningfulness in daily life. Content analysis was used for data analysis. RESULT Living with LGMD2 not only implies learning to live with the disease and the variations between good and bad periods but also means trying to make sense of a progressive disease that brings uncertainty about future health, by striving to make the best of the situation. Disease progression involves practical and mental struggle, trying to maintain control over one's life despite vanished physical functions that require continual adjustments to the body. Restrictions in a double sense were described, not only due to the disease but also due to poor comprehension of the disease in society. Lack of knowledge about LGMD2 among professionals often results in having to fight for the support needed. CONCLUSION In order to manage daily life, it is important to be seen and understood as an individual in contacts with professionals and in society in general, to have informal social support and meaningful activities as well as access to personal assistance if necessary. IMPLICATIONS FOR REHABILITATION Recessive limb-girdle muscular dystrophy (LGMD2) is a group of progressive disorders, which manifest in physical and psychological consequences for the individual. According to the salutogenic orientation, people need to find life comprehensible, manageable and meaningful, i.e. to achieve a sense of coherence (SOC), but living with LGMD2 may recurrently challenge the individual's SOC. Through the holistic view of the individual's situation that the salutogenic orientation provides, professionals may support the individual to strengthen SOC and thereby facilitate the movement towards health.
Collapse
Affiliation(s)
- Anna Carin Aho
- a Department of Health and Caring Sciences , Linnaeus University , Växjö , Sweden
| | - Sally Hultsjö
- b Psychiatric Clinic, County Hospital , Ryhov, Jönköping , Sweden , and
| | - Katarina Hjelm
- c Department of Social and Welfare Studies , Campus Norrköping, Linköping University , Linköping , Sweden
| |
Collapse
|
30
|
Zschüntzsch J, Jouvenal P, Zhang Y, Klinker F, Tiburcy M, Malzahn D, Liebetanz D, Brinkmeier H, Schmidt J. Human immunoglobulin G for experimental treatment of Duchenne muscular dystrophy. Clin Exp Immunol 2014; 178 Suppl 1:132-3. [PMID: 25546792 DOI: 10.1111/cei.12541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- J Zschüntzsch
- Clinic for Neurology, University of Göttingen, Göttingen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Theadom A, Rodrigues M, Roxburgh R, Balalla S, Higgins C, Bhattacharjee R, Jones K, Krishnamurthi R, Feigin V. Prevalence of muscular dystrophies: a systematic literature review. Neuroepidemiology 2014; 43:259-68. [PMID: 25532075 DOI: 10.1159/000369343] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Accepted: 10/19/2014] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Determining the prevalence of neuromuscular disorders for the general population is important to identify the scope of burden on society and enable comparisons with other health conditions. This systematic review aims to identify and collate the findings of studies published between 1960 and 2013 on the prevalence of all types of muscular dystrophies. SUMMARY Relevant articles were identified through electronic database searches and manual searches of reference lists. There were 38 articles from across 19 countries that met the inclusion criteria. The total combined prevalence for all muscular dystrophies for studies classified as having a low risk of bias ranged between 19.8 and 25.1 per 100,000 person-years. Myotonic dystrophy (0.5-18.1 per 100,000), Duchenne muscular dystrophy (1.7-4.2) and facioscapulohumeral muscular dystrophy (3.2-4.6 per 100,000) were found to be the most common types of disorder. There was wide variation in study methodology, case ascertainment, and verification procedures and populations studied, all of which may contribute to the wide prevalence range, in addition to the likely variation in prevalence by country. Key Messages: Greater consistency in the conduct and reporting of neuroepidemiological studies is urgently needed to enable comparisons to be made between studies, countries, and over time.
Collapse
Affiliation(s)
- Alice Theadom
- National Institute for Stroke and Applied Neuroscience, Auckland University of Technology, Auckland, New Zealand
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Wilton SD, Fletcher S, Flanigan KM. Dystrophin as a therapeutic biomarker: Are we ignoring data from the past? Neuromuscul Disord 2014; 24:463-6. [DOI: 10.1016/j.nmd.2014.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 03/11/2014] [Accepted: 03/13/2014] [Indexed: 01/16/2023]
|
33
|
Falcone G, Perfetti A, Cardinali B, Martelli F. Noncoding RNAs: emerging players in muscular dystrophies. BIOMED RESEARCH INTERNATIONAL 2014; 2014:503634. [PMID: 24729974 PMCID: PMC3960514 DOI: 10.1155/2014/503634] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Accepted: 01/17/2014] [Indexed: 12/16/2022]
Abstract
The fascinating world of noncoding RNAs has recently come to light, thanks to the development of powerful sequencing technologies, revealing a variety of RNA molecules playing important regulatory functions in most, if not all, cellular processes. Many noncoding RNAs have been implicated in regulatory networks that are determinant for skeletal muscle differentiation and disease. In this review, we outline the noncoding RNAs involved in physiological mechanisms of myogenesis and those that appear dysregulated in muscle dystrophies, also discussing their potential use as disease biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Germana Falcone
- Institute of Cell Biology and Neurobiology, National Research Council, 00015 Monterotondo Scalo, Italy
| | - Alessandra Perfetti
- Policlinico San Donato-IRCCS, Molecular Cardiology Laboratory, 20097 San Donato Milanese, Milan, Italy
| | - Beatrice Cardinali
- Institute of Cell Biology and Neurobiology, National Research Council, 00015 Monterotondo Scalo, Italy
| | - Fabio Martelli
- Policlinico San Donato-IRCCS, Molecular Cardiology Laboratory, 20097 San Donato Milanese, Milan, Italy
| |
Collapse
|
34
|
Sancisi V, Germinario E, Esposito A, Morini E, Peron S, Moggio M, Tomelleri G, Danieli-Betto D, Tupler R. Altered Tnnt3 characterizes selective weakness of fast fibers in mice overexpressing FSHD region gene 1 (FRG1). Am J Physiol Regul Integr Comp Physiol 2013; 306:R124-37. [PMID: 24305066 DOI: 10.1152/ajpregu.00379.2013] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD), a common hereditary myopathy, is characterized by atrophy and weakness of selective muscle groups. FSHD is considered an autosomal dominant disease with incomplete penetrance and unpredictable variability of clinical expression within families. Mice overexpressing FRG1 (FSHD region gene 1), a candidate gene for this disease, develop a progressive myopathy with features of the human disorder. Here, we show that in FRG1-overexpressing mice, fast muscles, which are the most affected by the dystrophic process, display anomalous fast skeletal troponin T (fTnT) isoform, resulting from the aberrant splicing of the Tnnt3 mRNA that precedes the appearance of dystrophic signs. We determine that muscles of FRG1 mice develop less strength due to impaired contractile properties of fast-twitch fibers associated with an anomalous MyHC-actin ratio and a reduced sensitivity to Ca(2+). We demonstrate that the decrease of Ca(2+) sensitivity of fast-twitch fibers depends on the anomalous troponin complex and can be rescued by the substitution with the wild-type proteins. Finally, we find that the presence of aberrant splicing isoforms of TNNT3 characterizes dystrophic muscles in FSHD patients. Collectively, our results suggest that anomalous TNNT3 profile correlates with the muscle impairment in both humans and mice. On the basis of these results, we propose that aberrant fTnT represents a biological marker of muscle phenotype severity and disease progression.
Collapse
Affiliation(s)
- Valentina Sancisi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Ca²⁺ plays a crucial role in connecting membrane excitability with contraction in myocardium. The hallmark features of heart failure are mechanical dysfunction and arrhythmias; defective intracellular Ca²⁺ homeostasis is a central cause of contractile dysfunction and arrhythmias in failing myocardium. Defective Ca²⁺ homeostasis in heart failure can result from pathological alteration in the expression and activity of an increasingly understood collection of Ca²⁺ homeostatic and structural proteins, ion channels, and enzymes. This review focuses on the molecular mechanisms of defective Ca²⁺ cycling in heart failure and considers how fundamental understanding of these pathways may translate into novel and innovative therapies.
Collapse
Affiliation(s)
- Min Luo
- Division of Cardiovascular Medicine, Department of Internal Medicine, Cardiovascular Research Center, Carver College of Medicine, University of Iowa, Iowa City, IA, USA
| | | |
Collapse
|
36
|
Wnt signaling in skeletal muscle dynamics: myogenesis, neuromuscular synapse and fibrosis. Mol Neurobiol 2013; 49:574-89. [PMID: 24014138 DOI: 10.1007/s12035-013-8540-5] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 08/15/2013] [Indexed: 12/21/2022]
Abstract
The signaling pathways activated by Wnt ligands are related to a wide range of critical cell functions, such as cell division, migration, and synaptogenesis. Here, we summarize compelling evidence on the role of Wnt signaling on several features of skeletal muscle physiology. We briefly review the role of Wnt pathways on the formation of muscle fibers during prenatal and postnatal myogenesis, highlighting its role on the activation of stem cells of the adult muscles. We also discuss how Wnt signaling regulates the precise formation of neuromuscular synapses, by modulating the differentiation of presynaptic and postsynaptic components, particularly regarding the clustering of acetylcholine receptors on the muscle membrane. In addition, based on previous evidence showing that Wnt pathways are linked to several diseases, such as Alzheimer's and cancer, we address recent studies indicating that Wnt signaling plays a key role in skeletal muscle fibrosis, a disease characterized by an increase in the extracellular matrix components leading to failure in muscle regeneration, tissue disorganization and loss of muscle activity. In this context, we also discuss the possible cross-talk between the Wnt/β-catenin pathway with two other critical profibrotic pathways, transforming growth factor β and connective tissue growth factor, which are potent stimulators of the accumulation of connective tissue, an effect characteristic of the fibrotic condition. As it has emerged in other pathological conditions, we suggests that muscle fibrosis may be a consequence of alterations of Wnt signaling activity.
Collapse
|
37
|
Murray JD, Canan BD, Martin CD, Stangland JE, Rastogi N, Rafael-Fortney JA, Janssen PML. The force-temperature relationship in healthy and dystrophic mouse diaphragm; implications for translational study design. Front Physiol 2012; 3:422. [PMID: 23162469 PMCID: PMC3491430 DOI: 10.3389/fphys.2012.00422] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2012] [Accepted: 10/17/2012] [Indexed: 11/21/2022] Open
Abstract
In the field of muscular dystrophy, striated muscle function is often assessed in vitro in dystrophin-deficient mdx mice in order to test the impact of a potential treatment strategy. Although many past studies have assessed diaphragm contractile function at or near room temperature, the diaphragm performs in vivo at 37°C. To improve translation of bench-top results to possible clinical application, we studied temperature-dependence of contractile performance in wild-type (C57BL/10) and mdx muscle strips at temperatures from 25°C to 37°C. Maximal tetanic force in wild-type muscles was higher at 37°C (198 ± 11 vs. 155 ± 9 mN/mm2 at 25°C), while the difference between wild-type and mdx was extremely similar: wild-type muscles produced 45.9% and 45.1% more force at 25°C and 37°C respectively. At 37°C twitch contraction kinetics and 50% rise time to tetanic plateau were slower in mdx diaphragm. A fatigue/injury protocol indicated 2-fold fatigue/contraction-induced force deficit in mdx muscles. We conclude that assessment of diaphragm muscle strips can be reliably and reproducibly performed at 37°C.
Collapse
Affiliation(s)
- Jason D Murray
- Department of Physiology and Cell Biology, The Ohio State University , Columbus, OH, USA
| | | | | | | | | | | | | |
Collapse
|
38
|
Ueyama M, Akimoto Y, Ichimiya T, Ueda R, Kawakami H, Aigaki T, Nishihara S. Increased apoptosis of myoblasts in Drosophila model for the Walker-Warburg syndrome. PLoS One 2010; 5:e11557. [PMID: 20644630 PMCID: PMC2903483 DOI: 10.1371/journal.pone.0011557] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2009] [Accepted: 06/17/2010] [Indexed: 11/18/2022] Open
Abstract
Walker-Warburg syndrome, a progressive muscular dystrophy, is a severe disease with various kinds of symptoms such as muscle weakness and occasional seizures. The genes of protein O-mannosyltransferases 1 and 2 (POMT1 and POMT2), fukutin, and fukutin-related protein are responsible for this syndrome. In our previous study, we cloned Drosophila orthologs of human POMT1 and POMT2 and identified their activity. However, the mechanism of onset of this syndrome is not well understood. Furthermore, little is known about the behavioral properties of the Drosophila POMT1 and POMT2 mutants, which are called rotated abdomen (rt) and twisted (tw), respectively. First, we performed various kinds of behavioral tests and described in detail the muscle structures by using these mutants. The mutant flies exhibited abnormalities in heavy exercises such as climbing or flight but not in light movements such as locomotion. Defective motor function in mutants appeared immediately after eclosion and was exaggerated with aging. Along with motor function, muscle ultrastructure in the tw mutant was altered, as seen in human patients. We demonstrated that expression of RNA interference (RNAi) for the rt gene and the tw mutant was almost completely lethal and semi-lethal, respectively. Flies expressing RNAi had reduced lifespans. These findings clearly demonstrate that Drosophila POMT mutants are models for human muscular dystrophy. We then observed a high density of myoblasts with an enhanced degree of apoptosis in the tw mutant, which completely lost enzymatic activity. In this paper, we propose a novel mechanism for the development of muscular dystrophy: POMT mutation causes high myoblast density and position derangement, which result in apoptosis, muscle disorganization, and muscle cell defects.
Collapse
Affiliation(s)
- Morio Ueyama
- Department of Bioinformatics, Soka University, Hachioji, Tokyo, Japan
| | - Yoshihiro Akimoto
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Tomomi Ichimiya
- Department of Bioinformatics, Soka University, Hachioji, Tokyo, Japan
| | - Ryu Ueda
- Invertebrate Genetics Laboratory, National Institute of Genetics, Mishima, Shizuoka, Japan
| | - Hayato Kawakami
- Department of Anatomy, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Toshiro Aigaki
- Department of Biological Science, Tokyo Metropolitan University, Hachioji, Tokyo, Japan
| | - Shoko Nishihara
- Department of Bioinformatics, Soka University, Hachioji, Tokyo, Japan
- * E-mail:
| |
Collapse
|
39
|
Proteomic profiling of x-linked muscular dystrophy. J Muscle Res Cell Motil 2010; 30:267-9. [DOI: 10.1007/s10974-009-9197-6] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Accepted: 12/24/2009] [Indexed: 01/10/2023]
|
40
|
Mitrpant C, Adams AM, Meloni PL, Muntoni F, Fletcher S, Wilton SD. Rational design of antisense oligomers to induce dystrophin exon skipping. Mol Ther 2009; 17:1418-26. [PMID: 19293776 PMCID: PMC2835229 DOI: 10.1038/mt.2009.49] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2008] [Accepted: 02/17/2009] [Indexed: 12/26/2022] Open
Abstract
Duchenne muscular dystrophy (DMD), one of the most severe neuromuscular disorders of childhood, is caused by the absence of a functional dystrophin. Antisense oligomer (AO) induced exon skipping is being investigated to restore functional dystrophin expression in models of muscular dystrophy and DMD patients. One of the major challenges will be in the development of clinically relevant oligomers and exon skipping strategies to address many different mutations. Various models, including cell-free extracts, cells transfected with artificial constructs, or mice with a human transgene, have been proposed as tools to facilitate oligomer design. Despite strong sequence homology between the human and mouse dystrophin genes, directing an oligomer to the same motifs in both species does not always induce comparable exon skipping. We report substantially different levels of exon skipping induced in normal and dystrophic human myogenic cell lines and propose that animal models or artificial assay systems useful in initial studies may be of limited relevance in designing the most efficient compounds to induce targeted skipping of human dystrophin exons for therapeutic outcomes.
Collapse
Affiliation(s)
- Chalermchai Mitrpant
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia, QE II Medical Centre, Nedlands, Western Australia, Australia
| | | | | | | | | | | |
Collapse
|
41
|
Mohri I, Aritake K, Taniguchi H, Sato Y, Kamauchi S, Nagata N, Maruyama T, Taniike M, Urade Y. Inhibition of prostaglandin D synthase suppresses muscular necrosis. THE AMERICAN JOURNAL OF PATHOLOGY 2009; 174:1735-44. [PMID: 19359520 DOI: 10.2353/ajpath.2009.080709] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Duchenne muscular dystrophy is a fatal muscle wasting disease that is characterized by a deficiency in the protein dystrophin. Previously, we reported that the expression of hematopoietic prostaglandin D synthase (HPGDS) appeared in necrotic muscle fibers from patients with either Duchenne muscular dystrophy or polymyositis. HPGDS is responsible for the production of the inflammatory mediator, prostaglandin D(2). In this paper, we validated the hypothesis that HPGDS has a role in the etiology of muscular necrosis. We investigated the expression of HPGDS/ prostaglandin D(2) signaling using two different mouse models of muscle necrosis, that is, bupivacaine-induced muscle necrosis and the mdx mouse, which has a genetic muscular dystrophy. We treated each mouse model with the HPGDS-specific inhibitor, HQL-79, and measured both necrotic muscle volume and selected cytokine mRNA levels. We confirmed that HPGDS expression was induced in necrotic muscle fibers in both bupivacaine-injected muscle and mdx mice. After administration of HQL-79, necrotic muscle volume was significantly decreased in both mouse models. Additionally, mRNA levels of both CD11b and transforming growth factor beta1 were significantly lower in HQL-79-treated mdx mice than in vehicle-treated animals. We also demonstrated that HQL-79 suppressed prostaglandin D(2) production and improved muscle strength in the mdx mouse. Our results show that HPGDS augments inflammation, which is followed by muscle injury. Furthermore, the inhibition of HPGDS ameliorates muscle necrosis even in cases of genetic muscular dystrophy.
Collapse
Affiliation(s)
- Ikuko Mohri
- Department of Molecular Behavioral Biology, Molecular Research Center for Child Mental Development, Osaka Bioscience Institute, Suita, Osaka, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Mitrpant C, Fletcher S, Iversen PL, Wilton SD. By-passing the nonsense mutation in the 4 CV mouse model of muscular dystrophy by induced exon skipping. J Gene Med 2009; 11:46-56. [PMID: 19006096 DOI: 10.1002/jgm.1265] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Duchenne muscular dystrophy (DMD), a severe neuromuscular disorder, is caused by protein-truncating mutations in the dystrophin gene. Absence of functional dystrophin renders muscle fibres more vulnerable to damage and necrosis. We report antisense oligomer (AO) induced exon skipping in the B6Ros.Cg-Dmd(mdx-4Cv)/J (4(CV)) mouse, a muscular dystrophy model arising from a nonsense mutation in dystrophin exon 53. Both exons 52 and 53 must be excised to remove the mutation and maintain the reading frame. METHODS A series of 2'-O-methyl modified oligomers on a phosphorothioate backbone (2OMeAOs) were designed and evaluated for the removal of each exon, and the most effective compounds were then combined to induce dual exon skipping in both myoblast cultures and in vivo. Exon skipping efficiency of 2OMeAOs and phosphorodiamidate morpholino oligomers (PMOs) was evaluated both in vitro and in vivo at the RNA and protein levels. RESULTS Compared to the original mdx mouse studies, induction of exon skipping from the 4(CV) dystrophin mRNA was far more challenging. PMO cocktails could restore synthesis of near-full length dystrophin protein in cultured 4(CV) myogenic cells and in vivo, after a single intramuscular injection. CONCLUSIONS By-passing the protein-truncating mutation in the 4(CV) mouse model of muscular dystrophy could not be achieved with single oligomers targeting both exons and was only achieved after the application of AO cocktails to remove exons 52 and 53. As in previous studies, the stability and efficiency of PMOs proved superior to 2OMeAOs for consistent and sustained protein induction in vivo.
Collapse
Affiliation(s)
- Chalermchai Mitrpant
- Centre for Neuromuscular and Neurological Disorders, University of Western Australia, QE II Medical Centre, Nedlands, Australia
| | | | | | | |
Collapse
|
43
|
Jazedje T, Secco M, Vieira NM, Zucconi E, Gollop TR, Vainzof M, Zatz M. Stem cells from umbilical cord blood do have myogenic potential, with and without differentiation induction in vitro. J Transl Med 2009; 7:6. [PMID: 19144182 PMCID: PMC2633316 DOI: 10.1186/1479-5876-7-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2008] [Accepted: 01/14/2009] [Indexed: 01/09/2023] Open
Abstract
The dystrophin gene, located at Xp21, codifies dystrophin, which is part of a protein complex responsible for the membrane stability of muscle cells. Its absence on muscle causes Duchenne Muscular Dystrophy (DMD), a severe disorder, while a defect of muscle dystrophin causes Becker Muscular Dystrophy (DMB), a milder disease. The replacement of the defective muscle through stem cells transplantation is a possible future treatment for these patients. Our objective was to analyze the potential of CD34+ stem cells from umbilical cord blood to differentiate in muscle cells and express dystrophin, in vitro. Protein expression was analyzed by Immunofluorescence, Western Blotting (WB) and Reverse Transcriptase – Polymerase Chain Reaction (RT-PCR). CD34+ stem cells and myoblasts from a DMD affected patient started to fuse with muscle cells immediately after co-cultures establishment. Differentiation in mature myotubes was observed after 15 days and dystrophin-positive regions were detected through Immunofluorescence analysis. However, WB or RT-PCR analysis did not detect the presence of normal dystrophin in co-cultures of CD34+ and DMD or DMB affected patients' muscle cells. In contrast, some CD34+ stem cells differentiated in dystrophin producers' muscle cells, what was observed by WB, reinforcing that this progenitor cell has the potential to originate muscle dystrophin in vitro, and not just in vivo like reported before.
Collapse
Affiliation(s)
- Tatiana Jazedje
- Department of Biology, Human Genome Research Center, São Paulo, Brazil.
| | | | | | | | | | | | | |
Collapse
|
44
|
Novel regulatory mechanisms for the proteoglycans decorin and biglycan during muscle formation and muscular dystrophy. Matrix Biol 2008; 27:700-8. [DOI: 10.1016/j.matbio.2008.07.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2008] [Revised: 06/24/2008] [Accepted: 07/01/2008] [Indexed: 01/11/2023]
|
45
|
Computer-aided visualization of muscle weakness distribution. J Neurol 2008; 255:1670-8. [DOI: 10.1007/s00415-008-0959-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2007] [Revised: 03/11/2008] [Accepted: 04/02/2008] [Indexed: 10/21/2022]
|
46
|
Molecular-targeted therapy for Duchenne muscular dystrophy: progress and potential. Mol Diagn Ther 2008; 12:99-108. [PMID: 18422374 DOI: 10.1007/bf03256275] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Duchenne muscular dystrophy (DMD) is a lethal heritable childhood myodegenerative condition caused by a mutation within the gene encoding the dystrophin protein within the X chromosome. While, historically, patients with this condition rarely lived into their thirties, they are now living substantially longer as a result of new treatments based on multi-disciplinary care. Despite these advances, the prognosis for DMD patients is limited, and a progressive reduction in quality of life and early death in adulthood cannot be prevented using currently available treatment regimens. The best hopes for a cure lies with cellular and gene therapy approaches that target the underlying genetic defect. In the past several years, viral and nonviral gene therapy methodologies based on adeno-associated viruses, naked plasmid delivery, antisense oligonucleotides, and oligonucleotide-mediated gene editing have advanced to a high degree of sophistication, to the extent that research has moved from the laboratory setting to the clinic. Notwithstanding these accomplishments, shortcomings with each therapy remain, so more work is required to devise an appropriate therapeutic strategy for the management and eventual cure of this debilitating disease.
Collapse
|
47
|
Budowle SA, Gonzalez S, Budowle B, Eisenberg AJ, Grange RW. A novel SNaPshot assay to detect the mdx mutation. Muscle Nerve 2008; 37:731-5. [PMID: 18506711 DOI: 10.1002/mus.21027] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The mdx mouse is an animal model for Duchenne muscular dystrophy (DMD). In order to evaluate possible treatments and to carry out genetic studies, it is essential to distinguish between mice that carry the dystrophic (mutant) or wild-type (wt) allele(s). The current amplification-resistant mutation system (ARMS) assay is labor intensive and yields false negatives, which reduces its efficiency as a screening tool. An alternate assay based on single-nucleotide polymorphism (SNP) primer extension technology (i.e., SNaPshot) is described. The SNaPshot assay has been optimized to identify both wild-type and mutant alleles, providing a robust, potentially automatable assay for high-throughput analysis.
Collapse
Affiliation(s)
- Sarah A Budowle
- Department of Human Nutrition, Food and Exercise, Virginia Polytechnic Institute and State University, Blacksburg, Virginia 24061, USA.
| | | | | | | | | |
Collapse
|
48
|
Resistance to infection and the muscular dystrophies--is there a molecular link? Neuromuscul Disord 2008; 18:423-5. [PMID: 18430573 DOI: 10.1016/j.nmd.2008.03.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2008] [Revised: 03/03/2008] [Accepted: 03/04/2008] [Indexed: 11/19/2022]
|
49
|
Mezzano V, Cabrera D, Vial C, Brandan E. Constitutively activated dystrophic muscle fibroblasts show a paradoxical response to TGF-beta and CTGF/CCN2. J Cell Commun Signal 2008; 1:205-17. [PMID: 18600480 DOI: 10.1007/s12079-008-0018-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Accepted: 02/09/2008] [Indexed: 12/20/2022] Open
Abstract
Transforming growth factor beta (TGF-beta) and connective tissue growth factor (CTGF) have been described to induce the production of extracellular matrix (ECM) proteins and have been reported to be increased in different fibrotic disorders. Skeletal muscle fibrosis is a common feature of Duchenne muscular dystrophy (DMD). The mdx mouse diaphragm is a good model for DMD since it reproduces the muscle degenerative and fibrotic changes. Fibronectin (FN) and proteoglycans (PG) are some of the ECM proteins upregulated in dystrophic conditions. In view of understanding the fibrotic process involved in DMD we have isolated fibroblasts from dystrophic mdx diaphragms. Here we report that regardless of the absence of degenerative myofibers, adult mdx diaphragm fibroblasts show increased levels of FN and condroitin/dermatan sulfate PGs synthesis. Fibroblasts isolated from non fibrotic tissue, such as 1 week old mice diaphragms or skin, do not present elevated FN levels. Furthermore, mdx fibroblast conditioned media is able to stimulate FN synthesis in control fibroblasts. Autocrine TGF-beta signaling was unaltered in mdx cells. When control fibroblasts are exposed to TGF-beta and CTGF, FN increases as expected. Paradoxically, in mdx cells it decreases in a concentration dependent manner and this decrease is not due to a downregulation of FN synthesis. According to this data we hypothesize that a pathological environment is able to reprogram fibroblasts into an activated phenotype which can be maintained through generations.
Collapse
Affiliation(s)
- Valeria Mezzano
- Centro de Regulación Celular y Patología, Departamento de Biología Celular y Molecular, Facultad de Ciencias Biológicas, MIFAB, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | | | | |
Collapse
|
50
|
Saccone V, Palmieri M, Passamano L, Piluso G, Meroni G, Politano L, Nigro V. Mutations that impair interaction properties of TRIM32 associated with limb-girdle muscular dystrophy 2H. Hum Mutat 2008; 29:240-7. [PMID: 17994549 DOI: 10.1002/humu.20633] [Citation(s) in RCA: 75] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
TRIM32 belongs to a large family of proteins characterized by a tripartite motif, possibly involved in the ubiquitination process, acting as an E3 ligase. In addition, TRIM32 has six NHL repeats with putative interaction properties. A homozygous mutation at the third NHL repeat (D487N) has been found in patients with limb girdle muscular dystrophy 2H (LGMD2H). This mutation was only identified in the inbred Manitoba Hutterite or their descendants. Interestingly, a mutation in the B-box domain of TRIM32 cosegregates with Bardet-Biedl syndrome type 11 (BBS11). The signs of BBS11 include obesity, pigmentary and retinal malformations, diabetes, polydactyly, and no muscular dystrophy, suggesting an alternative disease mechanism. We aim to ascertain whether D487N is the only pathological LGMD2H allele, limited to Hutterites. We studied the TRIM32 gene in 310 LGMD patients with no mutations at the other known loci. We identified four patients with novel mutated alleles. Two mutations were homozygous and missing in controls. These mutations also clustered at the NHL domain, suggesting that a specific (interaction) property might be abolished in LGMD2H patients. No mutations were found at the B-box region where the BBS11 mutation is found. We tested TRIM32 and its mutants by yeast-two-hybrid assay, developing an interaction test to validate mutations. All LGMD2H mutants, but not the BBS11, lost their ability to self-interact. The interaction of TRIM32 mutants with E2N, a protein involved in the ubiquitination process, was similarly impaired. In conclusion, the mutations here reported may cause muscular dystrophy by affecting the interaction properties of TRIM32.
Collapse
|