51
|
Saada YB, Dib C, Lipinski M, Vassetzky YS. Genome- and Cell-Based Strategies in Therapy of Muscular Dystrophies. BIOCHEMISTRY (MOSCOW) 2017; 81:678-90. [PMID: 27449614 DOI: 10.1134/s000629791607004x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Muscular dystrophies are a group of heterogeneous genetic disorders characterized by progressive loss of skeletal muscle mass. Depending on the muscular dystrophy, the muscle weakness varies in degree of severity. The majority of myopathies are due to genetic events leading to a loss of function of key genes involved in muscle function. Although there is until now no curative treatment to stop the progression of most myopathies, a significant number of experimental gene- and cell-based strategies and approaches have been and are being tested in vitro and in animal models, aiming to restore gene function. Genome editing using programmable endonucleases is a powerful tool for modifying target genome sequences and has been extensively used over the last decade to correct in vitro genetic defects of many single-gene diseases. By inducing double-strand breaks (DSBs), the engineered endonucleases specifically target chosen sequences. These DSBs are spontaneously repaired either by homologous recombination in the presence of a sequence template, or by nonhomologous-end joining error prone repair. In this review, we highlight recent developments and challenges for genome-editing based strategies that hold great promise for muscular dystrophies and regenerative medicine.
Collapse
Affiliation(s)
- Y Bou Saada
- UMR 8126, CNRS, Université Paris-Sud, Université Paris Saclay, Institut de Cancérologie Gustave-Roussy, Villejuif, F-94805, France.
| | | | | | | |
Collapse
|
52
|
Moyle LA, Blanc E, Jaka O, Prueller J, Banerji CR, Tedesco FS, Harridge SD, Knight RD, Zammit PS. Ret function in muscle stem cells points to tyrosine kinase inhibitor therapy for facioscapulohumeral muscular dystrophy. eLife 2016; 5. [PMID: 27841748 PMCID: PMC5108591 DOI: 10.7554/elife.11405] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 09/01/2016] [Indexed: 12/16/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) involves sporadic expression of DUX4, which inhibits myogenesis and is pro-apoptotic. To identify target genes, we over-expressed DUX4 in myoblasts and found that the receptor tyrosine kinase Ret was significantly up-regulated, suggesting a role in FSHD. RET is dynamically expressed during myogenic progression in mouse and human myoblasts. Constitutive expression of either RET9 or RET51 increased myoblast proliferation, whereas siRNA-mediated knockdown of Ret induced myogenic differentiation. Suppressing RET activity using Sunitinib, a clinically-approved tyrosine kinase inhibitor, rescued differentiation in both DUX4-expressing murine myoblasts and in FSHD patient-derived myoblasts. Importantly, Sunitinib also increased engraftment and differentiation of FSHD myoblasts in regenerating mouse muscle. Thus, DUX4-mediated activation of Ret prevents myogenic differentiation and could contribute to FSHD pathology by preventing satellite cell-mediated repair. Rescue of DUX4-induced pathology by Sunitinib highlights the therapeutic potential of tyrosine kinase inhibitors for treatment of FSHD. DOI:http://dx.doi.org/10.7554/eLife.11405.001
Collapse
Affiliation(s)
- Louise A Moyle
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom.,Department of Cell and Developmental Biology, University College London, London, United Kingdom
| | - Eric Blanc
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom.,Core Unit Bioinformatics, Berlin Institute of Health, Berlin, Germany.,Institute of Pathology, Charite Universitatsmedizin Berlin, Berlin, Germany
| | - Oihane Jaka
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, United Kingdom
| | - Johanna Prueller
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | - Christopher Rs Banerji
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| | | | - Stephen Dr Harridge
- Centre of Human and Aerospace Physiological Sciences, King's College London, London, United Kingdom
| | - Robert D Knight
- Craniofacial Development and Stem Cell Biology, King's College London, London, United Kingdom
| | - Peter S Zammit
- Randall Division of Cell and Molecular Biophysics, King's College London, London, United Kingdom
| |
Collapse
|
53
|
Randolph ME, Phillips BL, Choo HJ, Vest KE, Vera Y, Pavlath GK. Pharyngeal Satellite Cells Undergo Myogenesis Under Basal Conditions and Are Required for Pharyngeal Muscle Maintenance. Stem Cells 2016; 33:3581-95. [PMID: 26178867 DOI: 10.1002/stem.2098] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/15/2015] [Accepted: 06/01/2015] [Indexed: 12/19/2022]
Abstract
The pharyngeal muscles of the nasal, oral, and laryngeal pharynxes are required for swallowing. Pharyngeal muscles are preferentially affected in some muscular dystrophies yet spared in others. Muscle stem cells, called satellite cells, may be critical factors in the development of pharyngeal muscle disorders; however, very little is known about pharyngeal satellite cells (PSC) and their role in pharyngeal muscles. We show that PSC are distinct from the commonly studied hindlimb satellite cells both transcriptionally and biologically. Under basal conditions PSC proliferate, progress through myogenesis, and fuse with pharyngeal myofibers. Furthermore, PSC exhibit biologic differences dependent on anatomic location in the pharynx. Importantly, PSC are required to maintain myofiber size and myonuclear number in pharyngeal myofibers. Together, these results demonstrate that PSC are critical for pharyngeal muscle maintenance and suggest that satellite cell impairment could contribute to pharyngeal muscle pathology associated with various muscular dystrophies and aging.
Collapse
Affiliation(s)
| | | | - Hyo-Jung Choo
- Department of Pharmacology, Emory University, Atlanta, Georgia, USA
| | - Katherine E Vest
- Department of Pharmacology, Emory University, Atlanta, Georgia, USA
| | - Yandery Vera
- Department of Pharmacology, Emory University, Atlanta, Georgia, USA
| | - Grace K Pavlath
- Department of Pharmacology, Emory University, Atlanta, Georgia, USA
| |
Collapse
|
54
|
Knopp P, Krom YD, Banerji CRS, Panamarova M, Moyle LA, den Hamer B, van der Maarel SM, Zammit PS. DUX4 induces a transcriptome more characteristic of a less-differentiated cell state and inhibits myogenesis. J Cell Sci 2016; 129:3816-3831. [PMID: 27744317 PMCID: PMC5087662 DOI: 10.1242/jcs.180372] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Accepted: 08/13/2016] [Indexed: 12/14/2022] Open
Abstract
Skeletal muscle wasting in facioscapulohumeral muscular dystrophy (FSHD) results in substantial morbidity. On a disease-permissive chromosome 4qA haplotype, genomic and/or epigenetic changes at the D4Z4 macrosatellite repeat allows transcription of the DUX4 retrogene. Analysing transgenic mice carrying a human D4Z4 genomic locus from an FSHD-affected individual showed that DUX4 was transiently induced in myoblasts during skeletal muscle regeneration. Centromeric to the D4Z4 repeats is an inverted D4Z4 unit encoding DUX4c. Expression of DUX4, DUX4c and DUX4 constructs, including constitutively active, dominant-negative and truncated versions, revealed that DUX4 activates target genes to inhibit proliferation and differentiation of satellite cells, but that it also downregulates target genes to suppress myogenic differentiation. These transcriptional changes elicited by DUX4 in mouse have significant overlap with genes regulated by DUX4 in man. Comparison of DUX4 and DUX4c transcriptional perturbations revealed that DUX4 regulates genes involved in cell proliferation, whereas DUX4c regulates genes engaged in angiogenesis and muscle development, with both DUX4 and DUX4c modifing genes involved in urogenital development. Transcriptomic analysis showed that DUX4 operates through both target gene activation and repression to orchestrate a transcriptome characteristic of a less-differentiated cell state.
Collapse
Affiliation(s)
- Paul Knopp
- Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Yvonne D Krom
- Department of Human Genetics, Leiden University Medical Center, Leiden, Postbus 9600, 2300 RC, The Netherlands
| | - Christopher R S Banerji
- Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK Centre of Mathematics and Physics in the Life Sciences and Experimental Biology, University College London, London WC1E 6BT, UK
| | - Maryna Panamarova
- Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Louise A Moyle
- Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK
| | - Bianca den Hamer
- Department of Human Genetics, Leiden University Medical Center, Leiden, Postbus 9600, 2300 RC, The Netherlands
| | - Silvère M van der Maarel
- Department of Human Genetics, Leiden University Medical Center, Leiden, Postbus 9600, 2300 RC, The Netherlands
| | - Peter S Zammit
- Randall Division of Cell and Molecular Biophysics, Faculty of Life Sciences and Medicine, New Hunt's House, King's College London, Guy's Campus, London SE1 1UL, UK
| |
Collapse
|
55
|
Dmitriev P, Bou Saada Y, Dib C, Ansseau E, Barat A, Hamade A, Dessen P, Robert T, Lazar V, Louzada RAN, Dupuy C, Zakharova V, Carnac G, Lipinski M, Vassetzky YS. DUX4-induced constitutive DNA damage and oxidative stress contribute to aberrant differentiation of myoblasts from FSHD patients. Free Radic Biol Med 2016; 99:244-258. [PMID: 27519269 DOI: 10.1016/j.freeradbiomed.2016.08.007] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 08/04/2016] [Accepted: 08/08/2016] [Indexed: 12/29/2022]
Abstract
Facioscapulohumeral dystrophy (FSHD) is one of the three most common muscular dystrophies in the Western world, however, its etiology remains only partially understood. Here, we provide evidence of constitutive DNA damage in in vitro cultured myoblasts isolated from FSHD patients and demonstrate oxidative DNA damage implication in the differentiation of these cells into phenotypically-aberrant myotubes. Double homeobox 4 (DUX4), the major actor in FSHD pathology induced DNA damage accumulation when overexpressed in normal human myoblasts, and RNAi-mediated DUX4 inhibition reduced the level of DNA damage in FSHD myoblasts. Addition of tempol, a powerful antioxidant, to the culture medium of proliferating DUX4-transfected myoblasts and FSHD myoblasts reduced the level of DNA damage, suggesting that DNA alterations are mainly due to oxidative stress. Antioxidant treatment during the myogenic differentiation of FSHD myoblasts significantly reduced morphological defects in myotube formation. We propose that the induction of DNA damage is a novel function of the DUX4 protein affecting myogenic differentiation of FSHD myoblasts.
Collapse
Affiliation(s)
- Petr Dmitriev
- UMR 8126, Univ. Paris-Sud, CNRS, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France; PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, F-34295 Montpellier cedex 5, France
| | - Yara Bou Saada
- UMR 8126, Univ. Paris-Sud, CNRS, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France
| | - Carla Dib
- UMR 8126, Univ. Paris-Sud, CNRS, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France
| | - Eugénie Ansseau
- Laboratory of Molecular Biology, University of Mons, 20 place du Parc, B700 Mons, Belgium
| | - Ana Barat
- UMR 8126, Univ. Paris-Sud, CNRS, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France
| | - Aline Hamade
- ER030-EDST, Department of Life and Earth Sciences, Faculty of Sciences II, Lebanese University, Lebanon
| | - Philippe Dessen
- Functional Genomics Unit, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France
| | - Thomas Robert
- Functional Genomics Unit, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France
| | - Vladimir Lazar
- Functional Genomics Unit, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France
| | - Ruy A N Louzada
- UMR 8200, Univ., Paris-Sud, CNRS, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France
| | - Corinne Dupuy
- UMR 8200, Univ., Paris-Sud, CNRS, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France
| | - Vlada Zakharova
- Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, 119991 Moscow, Russia
| | - Gilles Carnac
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, F-34295 Montpellier cedex 5, France
| | - Marc Lipinski
- UMR 8126, Univ. Paris-Sud, CNRS, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France
| | - Yegor S Vassetzky
- UMR 8126, Univ. Paris-Sud, CNRS, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France; Lomonosov Moscow State University, Faculty of Bioengineering and Bioinformatics, 119991 Moscow, Russia.
| |
Collapse
|
56
|
Gatica LV, Rosa AL. A complex interplay of genetic and epigenetic events leads to abnormal expression of the DUX4 gene in facioscapulohumeral muscular dystrophy. Neuromuscul Disord 2016; 26:844-852. [PMID: 27816329 DOI: 10.1016/j.nmd.2016.09.015] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2016] [Revised: 09/13/2016] [Accepted: 09/16/2016] [Indexed: 12/16/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD), a prevalent inherited human myopathy, develops following a complex interplay of genetic and epigenetic events. FSHD1, the more frequent genetic form, is associated with: (1) deletion of an integral number of 3.3 Kb (D4Z4) repeated elements at the chromosomal region 4q35, (2) a specific 4q35 subtelomeric haplotype denominated 4qA, and (3) decreased methylation of cytosines at the 4q35-linked D4Z4 units. FSHD2 is most often caused by mutations at the SMCHD1 (Structural Maintenance of Chromosomes Hinge Domain 1) gene, on chromosome 18p11.32. FSHD2 individuals also carry the 4qA haplotype and decreased methylation of D4Z4 cytosines. Each D4Z4 unit contains a copy of the retrotransposed gene DUX4 (double homeobox containing protein 4). DUX4 gene functionality was questioned in the past because of its pseudogene-like structure, its location on repetitive telomeric DNA sequences (i.e. junk DNA), and the elusive nature of both the DUX4 transcript and the encoded protein, DUX4. It is now known that DUX4 is a nuclear-located transcription factor, which is normally expressed in germinal tissues. Aberrant DUX4 expression triggers a deregulation cascade inhibiting muscle differentiation, sensitizing cells to oxidative stress, and inducing muscle atrophy. A unifying pathogenic model for FSHD emerged with the recognition that the FSHD-permissive 4qA haplotype corresponds to a polyadenylation signal that stabilizes the DUX4 mRNA, allowing the toxic protein DUX4 to be expressed. This working hypothesis for FSHD pathogenesis highlights the intrinsic epigenetic nature of the molecular mechanism underlying FSHD as well as the pathogenic pathway connecting FSHD1 and FSHD2. Pharmacological control of either DUX4 gene expression or the activity of the DUX4 protein constitutes current potential rational therapeutic approaches to treat FSHD.
Collapse
Affiliation(s)
| | - Alberto Luis Rosa
- Laboratorio de Biología Celular y Molecular, Fundación Allende, Argentina; Servicio de Genética Médica y Laboratorio Diagnóstico Biología Molecular, Sanatorio Allende, Córdoba, Argentina; Laboratorio de Genética y Biología Molecular, Facultad de Ciencias Químicas, Universidad Católica de Córdoba, Argentina.
| |
Collapse
|
57
|
Caron L, Kher D, Lee KL, McKernan R, Dumevska B, Hidalgo A, Li J, Yang H, Main H, Ferri G, Petek LM, Poellinger L, Miller DG, Gabellini D, Schmidt U. A Human Pluripotent Stem Cell Model of Facioscapulohumeral Muscular Dystrophy-Affected Skeletal Muscles. Stem Cells Transl Med 2016; 5:1145-61. [PMID: 27217344 PMCID: PMC4996435 DOI: 10.5966/sctm.2015-0224] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 04/07/2016] [Indexed: 01/13/2023] Open
Abstract
UNLABELLED : Facioscapulohumeral muscular dystrophy (FSHD) represents a major unmet clinical need arising from the progressive weakness and atrophy of skeletal muscles. The dearth of adequate experimental models has severely hampered our understanding of the disease. To date, no treatment is available for FSHD. Human embryonic stem cells (hESCs) potentially represent a renewable source of skeletal muscle cells (SkMCs) and provide an alternative to invasive patient biopsies. We developed a scalable monolayer system to differentiate hESCs into mature SkMCs within 26 days, without cell sorting or genetic manipulation. Here we show that SkMCs derived from FSHD1-affected hESC lines exclusively express the FSHD pathogenic marker double homeobox 4 and exhibit some of the defects reported in FSHD. FSHD1 myotubes are thinner when compared with unaffected and Becker muscular dystrophy myotubes, and differentially regulate genes involved in cell cycle control, oxidative stress response, and cell adhesion. This cellular model will be a powerful tool for studying FSHD and will ultimately assist in the development of effective treatments for muscular dystrophies. SIGNIFICANCE This work describes an efficient and highly scalable monolayer system to differentiate human pluripotent stem cells (hPSCs) into skeletal muscle cells (SkMCs) and demonstrates disease-specific phenotypes in SkMCs derived from both embryonic and induced hPSCs affected with facioscapulohumeral muscular dystrophy. This study represents the first human stem cell-based cellular model for a muscular dystrophy that is suitable for high-throughput screening and drug development.
Collapse
Affiliation(s)
- Leslie Caron
- Genea Biocells Pty. Ltd., Sydney, New South Wales, Australia
| | - Devaki Kher
- Genea Biocells Pty. Ltd., Sydney, New South Wales, Australia
| | - Kian Leong Lee
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Robert McKernan
- Genea Biocells Pty. Ltd., Sydney, New South Wales, Australia
| | | | | | - Jia Li
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Henry Yang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Heather Main
- Genea Biocells Pty. Ltd., Sydney, New South Wales, Australia
| | - Giulia Ferri
- Dulbecco Telethon Institute and Division of Regenerative Medicine, San Raffaele Scientific Institute, Milano, Italy Vita-Salute San Raffaele University, Milano, Italy
| | - Lisa M Petek
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Lorenz Poellinger
- Cancer Science Institute of Singapore, National University of Singapore, Singapore
| | - Daniel G Miller
- Division of Genetic Medicine, Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Davide Gabellini
- Dulbecco Telethon Institute and Division of Regenerative Medicine, San Raffaele Scientific Institute, Milano, Italy
| | - Uli Schmidt
- Genea Biocells Pty. Ltd., Sydney, New South Wales, Australia Genea Biocells US Inc., San Diego, California, USA
| |
Collapse
|
58
|
Involvement of the FoxO1/MuRF1/Atrogin-1 Signaling Pathway in the Oxidative Stress-Induced Atrophy of Cultured Chronic Obstructive Pulmonary Disease Myotubes. PLoS One 2016; 11:e0160092. [PMID: 27526027 PMCID: PMC4987766 DOI: 10.1371/journal.pone.0160092] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Accepted: 07/13/2016] [Indexed: 12/16/2022] Open
Abstract
Oxidative stress is thought to be one of the most important mechanisms implicated in the muscle wasting of chronic obstructive pulmonary disease (COPD) patients, but its role has never been demonstrated. We therefore assessed the effects of both pro-oxidant and antioxidant treatments on the oxidative stress levels and atrophic signaling pathway of cultured COPD myotubes. Treatment of cultured COPD myotubes with the pro-oxidant molecule H2O2 resulted in increased ROS production (P = 0.002) and protein carbonylation (P = 0.050), in association with a more pronounced atrophy of the myotubes, as reflected by a reduced diameter (P = 0.003), and the activated expression of atrophic markers MuRF1 and FoxO1 (P = 0.022 and P = 0.030, respectively). Conversely, the antioxidant molecule ascorbic acid induced a reduction in ROS production (P<0.001) and protein carbonylation (P = 0.019), and an increase in the myotube diameter (P<0.001) to a level similar to the diameter of healthy subject myotubes, in association with decreased expression levels of MuRF1, atrogin-1 and FoxO1 (P<0.001, P = 0.002 and P = 0.042, respectively). A significant negative correlation was observed between the variations in myotube diameter and the variations in the expression of MuRF1 after antioxidant treatment (P = 0.047). Moreover, ascorbic acid was able to prevent the H2O2-induced atrophy of COPD myotubes. Last, the proteasome inhibitor MG132 restored the basal atrophy level of the COPD myotubes and also suppressed the H2O2-induced myotube atrophy. These findings demonstrate for the first time the involvement of oxidative stress in the atrophy of COPD peripheral muscle cells in vitro, via the FoxO1/MuRF1/atrogin-1 signaling pathway of the ubiquitin/proteasome system.
Collapse
|
59
|
Sente T, Van Berendoncks AM, Jonckheere AI, Rodenburg RJ, Lauwers P, Van Hoof V, Wouters A, Lardon F, Hoymans VY, Vrints CJ. Primary skeletal muscle myoblasts from chronic heart failure patients exhibit loss of anti-inflammatory and proliferative activity. BMC Cardiovasc Disord 2016; 16:107. [PMID: 27228977 PMCID: PMC4880810 DOI: 10.1186/s12872-016-0278-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Accepted: 05/13/2016] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Peripheral skeletal muscle wasting is a common finding with adverse effects in chronic heart failure (HF). Whereas its clinical relevance is beyond doubt, the underlying pathophysiological mechanisms are not yet fully elucidated. We aimed to introduce and characterize the primary culture of skeletal muscle cells from individual HF patients as a supportive model to study this muscle loss. METHODS AND RESULTS Primary myoblast and myotubes cultures were successfully propagated from the m. vastus lateralis of 6 HF patients with reduced ejection fraction (HFrEF; LVEF <45 %) and 6 age and gender-matched healthy donors. HFrEF cultures were not different from healthy donors in terms of morphology, such as myoblast size, shape and actin microfilament. Differentiation and fusion indexes were identical between groups. Myoblast proliferation in logarithmic growth phase, however, was attenuated in the HFrEF group (p = 0.032). In addition, HFrEF myoblasts are characterized by a reduced TNFR2 expression and IL-6 secretion (p = 0.017 and p = 0.016; respectively). CONCLUSION Biopsy derived primary skeletal muscle myoblasts of HFrEF patients produce similar morphological and myogenic differentiation responses as myoblasts of healthy donors, though demonstrate loss of anti-inflammatory and proliferative activity.
Collapse
Affiliation(s)
- Tahnee Sente
- Laboratory of Cellular and Molecular Cardiology, Antwerp University Hospital, Edegem, Belgium. .,Department of Translational Pathophysiological Research, Cardiovascular Diseases, University of Antwerp, Wilrijk, Belgium.
| | | | - An I Jonckheere
- Department of Pediatric Neurology, UZ Brussel, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Richard J Rodenburg
- Department of Pediatrics, Nijmegen Center for Mitochondrial Disorders, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Patrick Lauwers
- Department of Thoracic and Vascular Surgery, Antwerp University Hospital, Edegem, Belgium
| | - Viviane Van Hoof
- Department of Biochemistry, Antwerp University Hospital, Edegem, Belgium
| | - An Wouters
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Filip Lardon
- Center for Oncological Research (CORE) Antwerp, University of Antwerp, Wilrijk, Belgium
| | - Vicky Y Hoymans
- Laboratory of Cellular and Molecular Cardiology, Antwerp University Hospital, Edegem, Belgium.,Department of Cardiology, Antwerp University Hospital, Edegem, Belgium
| | - Christiaan J Vrints
- Laboratory of Cellular and Molecular Cardiology, Antwerp University Hospital, Edegem, Belgium.,Department of Translational Pathophysiological Research, Cardiovascular Diseases, University of Antwerp, Wilrijk, Belgium.,Department of Cardiology, Antwerp University Hospital, Edegem, Belgium
| |
Collapse
|
60
|
Hosford-Donovan A, Nilsson A, Wåhlin-Larsson B, Kadi F. Observational and mechanistic links between C-reactive protein and blood pressure in elderly women. Maturitas 2016; 89:52-7. [PMID: 27180160 DOI: 10.1016/j.maturitas.2016.04.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2015] [Revised: 04/09/2016] [Accepted: 04/18/2016] [Indexed: 11/17/2022]
Abstract
It is hypothesized that chronic systemic inflammation contributes to the age-related decline in cardiovascular function. The aim of the present study was to combine an assessment of the relationship between the serum level of C-reactive protein (CRP) and systolic and diastolic blood pressure in 108 elderly women (65 and 70 years) with an in-vitro exploration of the effects of CRP on the proliferative and angiogenic potential of endothelial cells exposed to serum in elderly women. Based on the median CRP level in our population, LowCRP (CRP<1.3mg/L) and HighCRP (>1.3mg/L) groups were identified. Body mass index, waist circumference, systolic blood pressure (SBP) and diastolic blood pressure (DBP) were significantly higher in the HighCRP group than in the LowCRP group (p<0.05). The influence of CRP on SBP and DBP remained significant after adjustments for BMI and use of antihypertensive medication (p<0.05). When adjusting for waist circumference the observed influence of CRP on SPB was attenuated (p=0.062). We next evaluated the ability to form capillary tubes (angiogenesis assay) and the proliferation rate of endothelial cells exposed to the sera of elderly women. Increased serum CRP levels were associated with an increased doubling time of endothelial cells (R(2)=0.39; p<0.05) and decreased capillary tube length (R(2)=0.30; p<0.05), indicating a reduction in the proliferation rate of endothelial cells and angiogenic potential. In conclusion, chronic inflammation influences blood pressure in elderly women and compromises endothelial cell function, thus contributing to the age-related decline in vascular health.
Collapse
Affiliation(s)
| | - Andreas Nilsson
- School of Health and Medical Sciences, Örebro University, 70182, Örebro, Sweden
| | | | - Fawzi Kadi
- School of Health and Medical Sciences, Örebro University, 70182, Örebro, Sweden.
| |
Collapse
|
61
|
Bou Saada Y, Dib C, Dmitriev P, Hamade A, Carnac G, Laoudj-Chenivesse D, Lipinski M, Vassetzky YS. Facioscapulohumeral dystrophy myoblasts efficiently repair moderate levels of oxidative DNA damage. Histochem Cell Biol 2016; 145:475-83. [PMID: 26860865 DOI: 10.1007/s00418-016-1410-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2016] [Indexed: 10/22/2022]
Abstract
Facioscapulohumeral dystrophy (FSHD) is a progressive muscular dystrophy linked to a deletion of a subset of D4Z4 macrosatellite repeats accompanied by a chromatin relaxation of the D4Z4 array on chromosome 4q. In vitro, FSHD primary myoblasts show altered expression of oxidative-related genes and are more susceptible to oxidative stress. Double homeobox 4 (DUX4) gene, encoded within each D4Z4 unit, is normally transcriptionally silenced but is found aberrantly expressed in skeletal muscles of FSHD patients. Its expression leads to a deregulation of DUX4 target genes including those implicated in redox balance. Here, we assessed DNA repair efficiency of oxidative DNA damage in FSHD myoblasts and DUX4-transfected myoblasts. We have shown that the DNA repair activity is altered neither in FSHD myoblasts nor in immortalized human myoblasts transiently expressing DUX4. DNA damage caused by moderate doses of an oxidant is efficiently repaired while FSHD myoblasts exposed for 24 h to high levels of oxidative stress accumulated more DNA damage than normal myoblasts, suggesting that FSHD myoblasts remain more vulnerable to oxidative stress at high doses of oxidants.
Collapse
Affiliation(s)
- Yara Bou Saada
- UMR 8126, CNRS, Univ. Paris-Sud, Institut de Cancérologie Gustave-Roussy, Université Paris Saclay, 94805, Villejuif, France
| | - Carla Dib
- UMR 8126, CNRS, Univ. Paris-Sud, Institut de Cancérologie Gustave-Roussy, Université Paris Saclay, 94805, Villejuif, France
| | - Petr Dmitriev
- UMR 8126, CNRS, Univ. Paris-Sud, Institut de Cancérologie Gustave-Roussy, Université Paris Saclay, 94805, Villejuif, France
| | - Aline Hamade
- ER030-EDST, Department of Life and Earth Sciences, Faculty of Sciences II, Lebanese University, Beirut, Lebanon
| | - Gilles Carnac
- INSERM U-1046, 371 Avenue du Doyen Gaston Giraud, 34295, Montpellier, France
| | | | - Marc Lipinski
- UMR 8126, CNRS, Univ. Paris-Sud, Institut de Cancérologie Gustave-Roussy, Université Paris Saclay, 94805, Villejuif, France
| | - Yegor S Vassetzky
- UMR 8126, CNRS, Univ. Paris-Sud, Institut de Cancérologie Gustave-Roussy, Université Paris Saclay, 94805, Villejuif, France. .,Koltzov Institute of Developmental Biology, Moscow, 117334, Russia.
| |
Collapse
|
62
|
Ansseau E, Eidahl JO, Lancelot C, Tassin A, Matteotti C, Yip C, Liu J, Leroy B, Hubeau C, Gerbaux C, Cloet S, Wauters A, Zorbo S, Meyer P, Pirson I, Laoudj-Chenivesse D, Wattiez R, Harper SQ, Belayew A, Coppée F. Homologous Transcription Factors DUX4 and DUX4c Associate with Cytoplasmic Proteins during Muscle Differentiation. PLoS One 2016; 11:e0146893. [PMID: 26816005 PMCID: PMC4729438 DOI: 10.1371/journal.pone.0146893] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2015] [Accepted: 12/24/2015] [Indexed: 12/26/2022] Open
Abstract
Hundreds of double homeobox (DUX) genes map within 3.3-kb repeated elements dispersed in the human genome and encode DNA-binding proteins. Among these, we identified DUX4, a potent transcription factor that causes facioscapulohumeral muscular dystrophy (FSHD). In the present study, we performed yeast two-hybrid screens and protein co-purifications with HaloTag-DUX fusions or GST-DUX4 pull-down to identify protein partners of DUX4, DUX4c (which is identical to DUX4 except for the end of the carboxyl terminal domain) and DUX1 (which is limited to the double homeodomain). Unexpectedly, we identified and validated (by co-immunoprecipitation, GST pull-down, co-immunofluorescence and in situ Proximal Ligation Assay) the interaction of DUX4, DUX4c and DUX1 with type III intermediate filament protein desmin in the cytoplasm and at the nuclear periphery. Desmin filaments link adjacent sarcomere at the Z-discs, connect them to sarcolemma proteins and interact with mitochondria. These intermediate filament also contact the nuclear lamina and contribute to positioning of the nuclei. Another Z-disc protein, LMCD1 that contains a LIM domain was also validated as a DUX4 partner. The functionality of DUX4 or DUX4c interactions with cytoplasmic proteins is underscored by the cytoplasmic detection of DUX4/DUX4c upon myoblast fusion. In addition, we identified and validated (by co-immunoprecipitation, co-immunofluorescence and in situ Proximal Ligation Assay) as DUX4/4c partners several RNA-binding proteins such as C1QBP, SRSF9, RBM3, FUS/TLS and SFPQ that are involved in mRNA splicing and translation. FUS and SFPQ are nuclear proteins, however their cytoplasmic translocation was reported in neuronal cells where they associated with ribonucleoparticles (RNPs). Several other validated or identified DUX4/DUX4c partners are also contained in mRNP granules, and the co-localizations with cytoplasmic DAPI-positive spots is in keeping with such an association. Large muscle RNPs were recently shown to exit the nucleus via a novel mechanism of nuclear envelope budding. Following DUX4 or DUX4c overexpression in muscle cell cultures, we observed their association with similar nuclear buds. In conclusion, our study demonstrated unexpected interactions of DUX4/4c with cytoplasmic proteins playing major roles during muscle differentiation. Further investigations are on-going to evaluate whether these interactions play roles during muscle regeneration as previously suggested for DUX4c.
Collapse
Affiliation(s)
- Eugénie Ansseau
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Jocelyn O. Eidahl
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America
| | - Céline Lancelot
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Alexandra Tassin
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Christel Matteotti
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Cassandre Yip
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Jian Liu
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America
| | - Baptiste Leroy
- Laboratory of Proteomic and Microbiology, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Céline Hubeau
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Cécile Gerbaux
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Samuel Cloet
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Armelle Wauters
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Sabrina Zorbo
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Pierre Meyer
- Pediatric Department, CHRU Montpellier, Montpellier, France
| | - Isabelle Pirson
- I.R.I.B.H.M., Free University of Brussels, Brussels, Belgium
| | | | - Ruddy Wattiez
- Laboratory of Proteomic and Microbiology, Research Institute for Biosciences, University of Mons, Mons, Belgium
| | - Scott Q. Harper
- Center for Gene Therapy, Research Institute at Nationwide Children's Hospital, Columbus, OH, United States of America
- Department of Pediatrics, Ohio State University College of Medicine, Columbus, OH, United States of America
| | - Alexandra Belayew
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Frédérique Coppée
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
- * E-mail:
| |
Collapse
|
63
|
Randolph ME, Pavlath GK. A muscle stem cell for every muscle: variability of satellite cell biology among different muscle groups. Front Aging Neurosci 2015; 7:190. [PMID: 26500547 PMCID: PMC4595652 DOI: 10.3389/fnagi.2015.00190] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2015] [Accepted: 09/21/2015] [Indexed: 12/22/2022] Open
Abstract
The human body contains approximately 640 individual skeletal muscles. Despite the fact that all of these muscles are composed of striated muscle tissue, the biology of these muscles and their associated muscle stem cell populations are quite diverse. Skeletal muscles are affected differentially by various muscular dystrophies (MDs), such that certain genetic mutations specifically alter muscle function in only a subset of muscles. Additionally, defective muscle stem cells have been implicated in the pathology of some MDs. The biology of muscle stem cells varies depending on the muscles with which they are associated. Here we review the biology of skeletal muscle stem cell populations of eight different muscle groups. Understanding the biological variation of skeletal muscles and their resident stem cells could provide valuable insight into mechanisms underlying the susceptibility of certain muscles to myopathic disease.
Collapse
|
64
|
Dib C, Saada YB, Dmitriev P, Richon C, Dessen P, Laoudj-Chenivesse D, Carnac G, Lipinski M, Vassetzky YS. Correction of the FSHD myoblast differentiation defect by fusion with healthy myoblasts. J Cell Physiol 2015. [DOI: 10.1002/jcp.25110] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Carla Dib
- UMR 8126, University of Paris-Sud, CNRS; Institut de Cancérologie Gustave-Roussy; F-94805 Villejuif France
| | - Yara Bou Saada
- UMR 8126, University of Paris-Sud, CNRS; Institut de Cancérologie Gustave-Roussy; F-94805 Villejuif France
| | - Petr Dmitriev
- UMR 8126, University of Paris-Sud, CNRS; Institut de Cancérologie Gustave-Roussy; F-94805 Villejuif France
| | - Catherine Richon
- Functional Genomics Unit; Institut de Cancérologie Gustave-Roussy; F-94805 Villejuif France
| | - Philippe Dessen
- Functional Genomics Unit; Institut de Cancérologie Gustave-Roussy; F-94805 Villejuif France
| | | | - Gilles Carnac
- INSERM U-1046; 371 Avenue du Doyen Gaston Giraud; F-34295 Montpellier France
| | - Marc Lipinski
- UMR 8126, University of Paris-Sud, CNRS; Institut de Cancérologie Gustave-Roussy; F-94805 Villejuif France
| | - Yegor S. Vassetzky
- UMR 8126, University of Paris-Sud, CNRS; Institut de Cancérologie Gustave-Roussy; F-94805 Villejuif France
| |
Collapse
|
65
|
Banerji CRS, Knopp P, Moyle LA, Severini S, Orrell RW, Teschendorff AE, Zammit PS. β-Catenin is central to DUX4-driven network rewiring in facioscapulohumeral muscular dystrophy. J R Soc Interface 2015; 12:20140797. [PMID: 25551153 DOI: 10.1098/rsif.2014.0797] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an incurable disease, characterized by skeletal muscle weakness and wasting. Genetically, FSHD is characterized by contraction or hypomethylation of repeat D4Z4 units on chromosome 4, which causes aberrant expression of the transcription factor DUX4 from the last repeat. Many genes have been implicated in FSHD pathophysiology, but an integrated molecular model is currently lacking. We developed a novel differential network methodology, Interactome Sparsification and Rewiring (InSpiRe), which detects network rewiring between phenotypes by integrating gene expression data with known protein interactions. Using InSpiRe, we performed a meta-analysis of multiple microarray datasets from FSHD muscle biopsies, then removed secondary rewiring using non-FSHD datasets, to construct a unified network of rewired interactions. Our analysis identified β-catenin as the main coordinator of FSHD-associated protein interaction signalling, with pathways including canonical Wnt, HIF1-α and TNF-α clearly perturbed. To detect transcriptional changes directly elicited by DUX4, gene expression profiling was performed using microarrays on murine myoblasts. This revealed that DUX4 significantly modified expression of the genes in our FSHD network. Furthermore, we experimentally confirmed that Wnt/β-catenin signalling is affected by DUX4 in murine myoblasts. Thus, we provide the first unified molecular map of FSHD signalling, capable of uncovering pathomechanisms and guiding therapeutic development.
Collapse
|
66
|
Kim E, Rich J, Karoutas A, Tarlykov P, Cochet E, Malysheva D, Mamchaoui K, Ogryzko V, Pirozhkova I. ZNF555 protein binds to transcriptional activator site of 4qA allele and ANT1: potential implication in Facioscapulohumeral dystrophy. Nucleic Acids Res 2015; 43:8227-42. [PMID: 26184877 PMCID: PMC4787827 DOI: 10.1093/nar/gkv721] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2013] [Accepted: 06/27/2015] [Indexed: 01/18/2023] Open
Abstract
Facioscapulohumeral dystrophy (FSHD) is an epi/genetic satellite disease associated with at least two satellite sequences in 4q35: (i) D4Z4 macrosatellite and (ii) β-satellite repeats (BSR), a prevalent part of the 4qA allele. Most of the recent FSHD studies have been focused on a DUX4 transcript inside D4Z4 and its tandem contraction in FSHD patients. However, the D4Z4-contraction alone is not pathological, which would also require the 4qA allele. Since little is known about BSR, we investigated the 4qA BSR functional role in the transcriptional control of the FSHD region 4q35. We have shown that an individual BSR possesses enhancer activity leading to activation of the Adenine Nucleotide Translocator 1 gene (ANT1), a major FSHD candidate gene. We have identified ZNF555, a previously uncharacterized protein, as a putative transcriptional factor highly expressed in human primary myoblasts that interacts with the BSR enhancer site and impacts the ANT1 promoter activity in FSHD myoblasts. The discovery of the functional role of the 4qA allele and ZNF555 in the transcriptional control of ANT1 advances our understanding of FSHD pathogenesis and provides potential therapeutic targets.
Collapse
Affiliation(s)
- Elena Kim
- CNRS, University Paris-Sud, UMR-8126, Gustave Roussy, Villejuif 94408, France
| | - Jeremy Rich
- CNRS, University Paris-Sud, UMR-8126, Gustave Roussy, Villejuif 94408, France
| | - Adam Karoutas
- CNRS, University Paris-Sud, UMR-8126, Gustave Roussy, Villejuif 94408, France
| | - Pavel Tarlykov
- National Center for Biotechnology, Astana 010000, Kazakhstan
| | - Emilie Cochet
- CNRS, University Paris-Sud, UMR-8126, Gustave Roussy, Villejuif 94408, France Proteomic Platform, IRCIV Gustave Roussy, Villejuif 94408, France
| | - Daria Malysheva
- CNRS, University Paris-Sud, UMR-8126, Gustave Roussy, Villejuif 94408, France
| | - Kamel Mamchaoui
- Thérapie des maladies du muscle strié, Institut de Myologie, UM76-Pierre et Marie CURIE University/U974-INSERM/UMR7215-CNRS, Paris 75013, France
| | - Vasily Ogryzko
- Proteomic Platform, IRCIV Gustave Roussy, Villejuif 94408, France INSERM, CNRS, University Paris-Sud, UMR-8126, Gustave Roussy, Villejuif 94408, France
| | - Iryna Pirozhkova
- CNRS, University Paris-Sud, UMR-8126, Gustave Roussy, Villejuif 94408, France
| |
Collapse
|
67
|
Caillaud C, Mechta M, Ainge H, Madsen AN, Ruell P, Mas E, Bisbal C, Mercier J, Twigg S, Mori TA, Simar D, Barrès R. Chronic erythropoietin treatment improves diet-induced glucose intolerance in rats. J Endocrinol 2015; 225:77-88. [PMID: 25767056 DOI: 10.1530/joe-15-0010] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/12/2015] [Indexed: 12/21/2022]
Abstract
Erythropoietin (EPO) ameliorates glucose metabolism through mechanisms not fully understood. In this study, we investigated the effect of EPO on glucose metabolism and insulin signaling in skeletal muscle. A 2-week EPO treatment of rats fed with a high-fat diet (HFD) improved fasting glucose levels and glucose tolerance, without altering total body weight or retroperitoneal fat mass. Concomitantly, EPO partially rescued insulin-stimulated AKT activation, reduced markers of oxidative stress, and restored heat-shock protein 72 expression in soleus muscles from HFD-fed rats. Incubation of skeletal muscle cell cultures with EPO failed to induce AKT phosphorylation and had no effect on glucose uptake or glycogen synthesis. We found that the EPO receptor gene was expressed in myotubes, but was undetectable in soleus. Together, our results indicate that EPO treatment improves glucose tolerance but does not directly activate the phosphorylation of AKT in muscle cells. We propose that the reduced systemic inflammation or oxidative stress that we observed after treatment with EPO could contribute to the improvement of whole-body glucose metabolism.
Collapse
Affiliation(s)
- Corinne Caillaud
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - Mie Mechta
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - Heidi Ainge
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - Andreas N Madsen
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, N
| | - Patricia Ruell
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - Emilie Mas
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - Catherine Bisbal
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - Jacques Mercier
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, N
| | - Stephen Twigg
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - Trevor A Mori
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - David Simar
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| | - Romain Barrès
- Exercise Health and Performance Faculty of Health Sciences, and Charles Perkins Centre, The University of Sydney, Sydney, New South Wales, Australia Faculty of Health and Medical Sciences The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark Department of Neuroscience and Pharmacology Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark School of Medicine and Pharmacology Royal Perth Hospital, The University of Western Australia, Perth, Western Australia, Australia UMR CNRS 9214 U1046 INSERM Physiologie et Médecine Expérimentale du Cœur et des Muscles, Université de Montpellier, Montpellier, France Physiology Department CHU Arnaud de Villeneuve, Montpellier, France Department of Endocrinology Sydney Medical School, Royal Prince Alfred Hospital, University of Sydney, Camperdown, New South Wales, Australia Inflammation and Infection Research School of Medical Sciences, UNSW Australia, Sydney, New South Wales, Australia
| |
Collapse
|
68
|
Passerieux E, Hayot M, Jaussent A, Carnac G, Gouzi F, Pillard F, Picot MC, Böcker K, Hugon G, Pincemail J, Defraigne JO, Verrips T, Mercier J, Laoudj-Chenivesse D. Effects of vitamin C, vitamin E, zinc gluconate, and selenomethionine supplementation on muscle function and oxidative stress biomarkers in patients with facioscapulohumeral dystrophy: a double-blind randomized controlled clinical trial. Free Radic Biol Med 2015; 81:158-69. [PMID: 25246239 DOI: 10.1016/j.freeradbiomed.2014.09.014] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2014] [Revised: 09/11/2014] [Accepted: 09/11/2014] [Indexed: 12/29/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disease characterized by progressive weakness and atrophy of specific skeletal muscles. As growing evidence suggests that oxidative stress may contribute to FSHD pathology, antioxidants that might modulate or delay oxidative insults could help in maintaining FSHD muscle function. Our primary objective was to test whether oral administration of vitamin C, vitamin E, zinc gluconate, and selenomethionine could improve the physical performance of patients with FSHD. Adult patients with FSHD (n=53) were enrolled at Montpellier University Hospital (France) in a randomized, double-blind, placebo-controlled pilot clinical trial. Patients were randomly assigned to receive 500 mg vitamin C, 400mg vitamin E, 25mg zinc gluconate and 200 μg selenomethionine (n=26), or matching placebo (n=27) once a day for 17 weeks. Primary outcomes were changes in the two-minute walking test (2-MWT), maximal voluntary contraction, and endurance limit time of the dominant and nondominant quadriceps (MVCQD, MVCQND, TlimQD, and TlimQND, respectively) after 17 weeks of treatment. Secondary outcomes were changes in the antioxidant status and oxidative stress markers. Although 2-MWT, MVCQ, and TlimQ were all significantly improved in the supplemented group at the end of the treatment compared to baseline, only MVCQ and TlimQ variations were significantly different between groups (MVCQD: P=0.011; MVCQND: P=0.004; TlimQD: P=0.028; TlimQND: P=0.011). Similarly, the vitamin C (P<0.001), vitamin E as α-tocopherol (P<0.001), vitamin C/vitamin E ratio (P=0.017), vitamin E γ/α ratio (P=0.022) and lipid peroxides (P<0.001) variations were significantly different between groups. In conclusion, vitamin E, vitamin C, zinc, and selenium supplementation has no significant effect on the 2-MWT, but improves MVCQ and TlimQ of both quadriceps by enhancing the antioxidant defenses and reducing oxidative stress. This trial was registered at clinicaltrials.gov (number: NCT01596803).
Collapse
Affiliation(s)
- Emilie Passerieux
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France
| | - Maurice Hayot
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France
| | - Audrey Jaussent
- Department of Biostatistics and Epidemiology, University Hospital of Montpellier, Montpellier, France
| | - Gilles Carnac
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France
| | - Fares Gouzi
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France
| | - Fabien Pillard
- Department of Respiratory Exploration and Department of Sports Medicine, Larrey University Hospital, Toulouse CEDEX, France
| | - Marie-Christine Picot
- Department of Biostatistics and Epidemiology, University Hospital, Montpellier, France and CIC 1001-INSERM
| | - Koen Böcker
- Alan Turing Institute Almere, The Netherlands
| | - Gerald Hugon
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France
| | - Joel Pincemail
- Department of cardiovascular Surgery and Department of CREDEC, University Hospital of Liege, Belgium
| | - Jean O Defraigne
- Department of cardiovascular Surgery and Department of CREDEC, University Hospital of Liege, Belgium
| | - Theo Verrips
- Utrecht University, Department of Biology, The Netherlands
| | - Jacques Mercier
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France
| | - Dalila Laoudj-Chenivesse
- University of Montpellier 1 and 2, INSERM Unit 1046, Montpellier, France and Department of Clinical Physiology, University Hospital, Montpellier, France.
| |
Collapse
|
69
|
Pomiès P, Rodriguez J, Blaquière M, Sedraoui S, Gouzi F, Carnac G, Laoudj-Chenivesse D, Mercier J, Préfaut C, Hayot M. Reduced myotube diameter, atrophic signalling and elevated oxidative stress in cultured satellite cells from COPD patients. J Cell Mol Med 2014; 19:175-86. [PMID: 25339614 PMCID: PMC4288361 DOI: 10.1111/jcmm.12390] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Accepted: 07/07/2014] [Indexed: 12/02/2022] Open
Abstract
The mechanisms leading to skeletal limb muscle dysfunction in chronic obstructive pulmonary disease (COPD) have not been fully elucidated. Exhausted muscle regenerative capacity of satellite cells has been evocated, but the capacity of satellite cells to proliferate and differentiate properly remains unknown. Our objectives were to compare the characteristics of satellite cells derived from COPD patients and healthy individuals, in terms of proliferative and differentiation capacities, morphological phenotype and atrophy/hypertrophy signalling, and oxidative stress status. Therefore, we purified and cultivated satellite cells from progressively frozen vastus lateralis biopsies of eight COPD patients and eight healthy individuals. We examined proliferation parameters, differentiation capacities, myotube diameter, expression of atrophy/hypertrophy markers, oxidative stress damages, antioxidant enzyme expression and cell susceptibility to H2O2 in cultured myoblasts and/or myotubes. Proliferation characteristics and commitment to terminal differentiation were similar in COPD patients and healthy individuals, despite impaired fusion capacities of COPD myotubes. Myotube diameter was smaller in COPD patients (P = 0.015), and was associated with a higher expression of myostatin (myoblasts: P = 0.083; myotubes: P = 0.050) and atrogin-1 (myoblasts: P = 0.050), and a decreased phospho-AKT/AKT ratio (myoblasts: P = 0.022). Protein carbonylation (myoblasts: P = 0.028; myotubes: P = 0.002) and lipid peroxidation (myotubes: P = 0.065) were higher in COPD cells, and COPD myoblasts were significantly more susceptible to oxidative stress. Thus, cultured satellite cells from COPD patients display characteristics of morphology, atrophic signalling and oxidative stress similar to those described in in vivo COPD skeletal limb muscles. We have therefore demonstrated that muscle alteration in COPD can be studied by classical in vitro cellular models.
Collapse
Affiliation(s)
- Pascal Pomiès
- INSERM U-1046, University Montpellier I, University Montpellier II, Montpellier, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
70
|
Wåhlin-Larsson B, Carnac G, Kadi F. The influence of systemic inflammation on skeletal muscle in physically active elderly women. AGE (DORDRECHT, NETHERLANDS) 2014; 36:9718. [PMID: 25311555 PMCID: PMC4199340 DOI: 10.1007/s11357-014-9718-0] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 10/06/2014] [Indexed: 06/04/2023]
Abstract
The biological mechanisms responsible for the decline in skeletal muscle mass during aging remain unknown. It is hypothesized that elevations in the level of the acute phase C-reactive protein (CRP) negatively affect skeletal muscle mass in elderly. We examined the relationship between serum CRP and muscle mass in a population of active elderly women (65-70 years; n =23). Though all subjects were physically active, serum CRP levels were negatively associated to the amount of time spent in moderate-to-vigorous physical activity (R(2) = 0.20, P = 0.032) and to skeletal muscle mass (R(2) = 0.28, P = 0.009). We further aimed to determine the potential mechanisms behind the action of systemic inflammation on skeletal muscle by exposing myoblasts isolated from vastus lateralis to the different sera from each elderly woman. The doubling time (DT) of myoblasts increased when cells were exposed to sera with high CRP levels (R(2) = 0.27, P = 0.011), indicating that CRP contributes to the impairment of the proliferative rate of myoblasts in elderly. In order to further confirm our findings, we incubated human myoblasts in exogenous CRP. Exposition to exogenous CRP induced an increase in myoblast DT by 1.21-fold (P = 0.007) and a reduction in the expression of the proliferation marker ki-67 confirming the negative influence of CRP on myoblast proliferative rate. Collectively, these findings highlight the contribution of the systemic inflammatory status in the age-related decline in skeletal muscle function.
Collapse
Affiliation(s)
- Britta Wåhlin-Larsson
- />School of Health and Medical Sciences, Örebro University, SE-701 82 Örebro, Sweden
| | - Gilles Carnac
- />INSERM U1046, University Montpellier I, Montpellier, France
| | - Fawzi Kadi
- />School of Health and Medical Sciences, Örebro University, SE-701 82 Örebro, Sweden
| |
Collapse
|
71
|
Defects in TLR3 expression and RNase L activation lead to decreased MnSOD expression and insulin resistance in muscle cells of obese people. Cell Death Dis 2014; 5:e1136. [PMID: 24651439 PMCID: PMC3973244 DOI: 10.1038/cddis.2014.104] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Revised: 02/12/2014] [Accepted: 02/13/2014] [Indexed: 12/14/2022]
Abstract
Obesity is associated with chronic low-grade inflammation and oxidative stress that blunt insulin response in its target tissues, leading to insulin resistance (IR). IR is a characteristic feature of type 2 diabetes. Skeletal muscle is responsible for 75% of total insulin-dependent glucose uptake; consequently, skeletal muscle IR is considered to be the primary defect of systemic IR development. Interestingly, some obese people stay insulin-sensitive and metabolically healthy. With the aim of understanding this difference and identifying the mechanisms responsible for insulin sensitivity maintenance/IR development during obesity, we explored the role of the latent endoribonuclease (RNase L) in skeletal muscle cells. RNase L is a regulator of innate immunity, of double-stranded RNA sensors and of toll-like receptor (TLR) 4 signaling. It is regulated during inflammation by interferons and its activity is dependent on its binding to 2-5A, an oligoadenylate synthesized by oligoadenylate synthetases (OAS). Increased expression of RNase L or downregulation of its inhibitor (RLI) improved insulin response in mouse myogenic C2C12 cells and in primary human myotubes from normal-weight subjects treated with palmitate, a saturated free fatty acid (FFA) known to induce inflammation and oxidative stress via TLR4 activation. While RNase L and RLI levels remained unchanged, OAS level was decreased in primary myotubes from insulin-resistant obese subjects (OB-IR) compared with myotubes from insulin-sensitive obese subjects (OB-IS). TLR3 and mitochondrial manganese superoxide dismutase (MnSOD) were also underexpressed in OB-IR myotubes. Activation of RNase L by 2-5A transfection allowed to restore insulin response, OAS, MnSOD and TLR3 expression in OB-IR myotubes. Due to low expression of OAS, OB-IR myotubes present a defect in RNase L activation and TLR3 regulation. Consequently, MnSOD level is low and insulin sensitivity is reduced. These results support that RNase L activity limits FFA/obesity-induced impairment of insulin response in muscle cells via TLR3 and MnSOD expression.
Collapse
|
72
|
Bachasson D, Temesi J, Bankole C, Lagrange E, Boutte C, Millet G, Verges S, Levy P, Feasson L, Wuyam B. Assessement of quadriceps strength, endurance and fatigue in FSHD and CMT: Benefits and limits of femoral nerve magnetic stimulation. Clin Neurophysiol 2014; 125:396-405. [DOI: 10.1016/j.clinph.2013.08.001] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 07/30/2013] [Accepted: 08/01/2013] [Indexed: 12/30/2022]
|
73
|
DNA polymorphism and epigenetic marks modulate the affinity of a scaffold/matrix attachment region to the nuclear matrix. Eur J Hum Genet 2014; 22:1117-23. [PMID: 24448543 DOI: 10.1038/ejhg.2013.306] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 12/05/2013] [Accepted: 12/11/2013] [Indexed: 12/22/2022] Open
Abstract
Mechanisms that regulate attachment of the scaffold/matrix attachment regions (S/MARs) to the nuclear matrix remain largely unknown. We have studied the effect of simple sequence length polymorphism (SSLP), DNA methylation and chromatin organization in an S/MAR implicated in facioscapulohumeral dystrophy (FSHD), a hereditary disease linked to a partial deletion of the D4Z4 repeat array on chromosome 4q. This FSHD-related nuclear matrix attachment region (FR-MAR) loses its efficiency in myoblasts from FSHD patients. Three criteria were found to be important for high-affinity interaction between the FR-MAR and the nuclear matrix: the presence of a specific SSLP haplotype in chromosomal DNA, the methylation of one specific CpG within the FR-MAR and the absence of histone H3 acetylated on lysine 9 in the relevant chromatin fragment.
Collapse
|
74
|
Dmitriev P, Kairov U, Robert T, Barat A, Lazar V, Carnac G, Laoudj-Chenivesse D, Vassetzky YS. Cancer-related genes in the transcription signature of facioscapulohumeral dystrophy myoblasts and myotubes. J Cell Mol Med 2013; 18:208-17. [PMID: 24341522 PMCID: PMC3930408 DOI: 10.1111/jcmm.12182] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 10/08/2013] [Indexed: 01/23/2023] Open
Abstract
Muscular dystrophy is a condition potentially predisposing for cancer; however, currently, only Myotonic dystrophy patients are known to have a higher risk of cancer. Here, we have searched for a link between facioscapulohumeral dystrophy (FSHD) and cancer by comparing published transcriptome signatures of FSHD and various malignant tumours and have found a significant enrichment of cancer-related genes among the genes differentially expressed in FSHD. The analysis has shown that gene expression profiles of FSHD myoblasts and myotubes resemble that of Ewing's sarcoma more than that of other cancer types tested. This is the first study demonstrating a similarity between FSHD and cancer cell expression profiles, a finding that might indicate the existence of a common step in the pathogenesis of these two diseases.
Collapse
Affiliation(s)
- Petr Dmitriev
- UMR8126, Université Paris-Sud 11, CNRS, Institut de cancérologie Gustave Roussy, Villejuif, France; INSERM U1046, Université Montpellier I, Montpellier, France
| | | | | | | | | | | | | | | |
Collapse
|
75
|
Dmitriev P, Stankevicins L, Ansseau E, Petrov A, Barat A, Dessen P, Robert T, Turki A, Lazar V, Labourer E, Belayew A, Carnac G, Laoudj-Chenivesse D, Lipinski M, Vassetzky YS. Defective regulation of microRNA target genes in myoblasts from facioscapulohumeral dystrophy patients. J Biol Chem 2013; 288:34989-5002. [PMID: 24145033 DOI: 10.1074/jbc.m113.504522] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant hereditary neuromuscular disorder linked to the deletion of an integral number of 3.3-kb-long macrosatellite repeats (D4Z4) within the subtelomeric region of chromosome 4q. Most genes identified in this region are overexpressed in FSHD myoblasts, including the double homeobox genes DUX4 and DUX4c. We have carried out a simultaneous miRNome/transcriptome analysis of FSHD and control primary myoblasts. Of 365 microRNAs (miRNAs) analyzed in this study, 29 were found to be differentially expressed between FSHD and normal myoblasts. Twenty-one microRNAs (miR-1, miR-7, miR-15a, miR-22, miR-30e, miR-32, miR-107, miR-133a, miR-133b, miR-139, miR-152, miR-206, miR-223, miR-302b, miR-331, miR-362, miR-365, miR-382, miR-496, miR-532, miR-654, and miR-660) were up-regulated, and eight were down-regulated (miR-15b, miR-20b, miR-21, miR-25, miR-100, miR-155, miR-345, and miR-594). Twelve of the miRNAs up-regulated in FHSD were also up-regulated in the cells ectopically expressing DUX4c, suggesting that this gene could regulate miRNA gene transcription. The myogenic miRNAs miR-1, miR-133a, miR-133b, and miR-206 were highly expressed in FSHD myoblasts, which nonetheless did not prematurely enter myogenic differentiation. This could be accounted for by the fact that in FSHD myoblasts, functionally important target genes, including cell cycle, DNA damage, and ubiquitination-related genes, escape myogenic microRNA-induced repression.
Collapse
Affiliation(s)
- Petr Dmitriev
- From UMR 8126, Université Paris-Sud, CNRS, Institut de Cancérologie Gustave-Roussy, F-94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
76
|
Hauerslev S, Ørngreen MC, Hertz JM, Vissing J, Krag TO. Muscle regeneration and inflammation in patients with facioscapulohumeral muscular dystrophy. Acta Neurol Scand 2013; 128:194-201. [PMID: 23410246 DOI: 10.1111/ane.12109] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2013] [Indexed: 01/02/2023]
Abstract
BACKGROUND AND OBJECTIVES The aim of this study was to investigate whether inflammation and regeneration are prominent in mildly affected muscles of patients with facioscapulohumeral muscular dystrophy type 1A (FSHD1A). Inflammation in muscle has been suggested by MRI studies in patients with FSHD1A. METHODS We analysed immunohistological and histological stains of muscle biopsies from 24 patients with FSHD1A, using 10 patients with Becker muscular dystrophy (BMD) for comparison. RESULTS Internalized nuclei were more prevalent in BMD (23.7 ± 10.8%) vs FSHD1A (6.3 ± 6.8%; P < 0.001), indicating more past regenerating fibres in BMD. Recently regenerating fibres, expressing neonatal myosin heavy chain and vimentin, did not differ significantly between patients with FSHD1A (1.1 ± 2.9%) and patients with BMD (1.8 ± 1.9%). Regeneration was not correlated with the number of KpnI restriction fragment repeats, an FSHD1A-defining genotype property within the D4Z4 locus, or overall disease severity in patients with FSHD1A. Macrophages were more prevalent in FSHD1A (0.50 ± 0.63 per mm(2) ) vs BMD (0.07 ± 0.07 per mm(2) ), whereas inflammatory T cells were equally infrequent. CONCLUSIONS Macrophages were more prevalent in patients with FSHD1A and could be an important pathogenic mechanism for the initiation of the dystrophic process. Furthermore, regeneration was unrelated to genotype and disease severity in FSHD1A.
Collapse
Affiliation(s)
- S. Hauerslev
- Neuromuscular Research Unit; Department of Neurology; Rigs hospitalet; University of Copenhagen; Copenhagen; Denmark
| | - M. C. Ørngreen
- Neuromuscular Research Unit; Department of Neurology; Rigs hospitalet; University of Copenhagen; Copenhagen; Denmark
| | - J. M. Hertz
- Department of Clinical Genetics; Odense University Hospital; Odense; Denmark
| | - J. Vissing
- Neuromuscular Research Unit; Department of Neurology; Rigs hospitalet; University of Copenhagen; Copenhagen; Denmark
| | - T. O. Krag
- Neuromuscular Research Unit; Department of Neurology; Rigs hospitalet; University of Copenhagen; Copenhagen; Denmark
| |
Collapse
|
77
|
Block GJ, Narayanan D, Amell AM, Petek LM, Davidson KC, Bird TD, Tawil R, Moon RT, Miller DG. Wnt/β-catenin signaling suppresses DUX4 expression and prevents apoptosis of FSHD muscle cells. Hum Mol Genet 2013; 22:4661-72. [PMID: 23821646 DOI: 10.1093/hmg/ddt314] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy is a dominantly inherited myopathy associated with chromatin relaxation of the D4Z4 macrosatellite array on chromosome 4. DUX4 is encoded within each unit of the D4Z4 array where it is normally transcriptionally silenced and packaged as constitutive heterochromatin. Truncation of the array to less than 11 D4Z4 units (FSHD1) or mutations in SMCHD1 (FSHD2) results in chromatin relaxation and a small percentage of cultured myoblasts from these individuals exhibit infrequent bursts of DUX4 expression. There are no cellular or animal models to determine the trigger of the DUX4 producing transcriptional bursts and there has been a failure to date to detect the protein in significant numbers of cells from FSHD-affected individuals. Here, we demonstrate for the first time that myotubes generated from FSHD patients express sufficient amounts of DUX4 to undergo DUX4-dependent apoptosis. We show that activation of the Wnt/β-catenin signaling pathway suppresses DUX4 transcription in FSHD1 and FSHD2 myotubes and can rescue DUX4-mediated myotube apoptosis. In addition, reduction of mRNA transcripts from Wnt pathway genes β-catenin, Wnt3A and Wnt9B results in DUX4 activation. We propose that Wnt/β-catenin signaling is important for transcriptional repression of DUX4 and identify a novel group of therapeutic targets for the treatment of FSHD.
Collapse
|
78
|
Neguembor MV, Xynos A, Onorati MC, Caccia R, Bortolanza S, Godio C, Pistoni M, Corona DF, Schotta G, Gabellini D. FSHD muscular dystrophy region gene 1 binds Suv4-20h1 histone methyltransferase and impairs myogenesis. J Mol Cell Biol 2013; 5:294-307. [DOI: 10.1093/jmcb/mjt018] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
79
|
Sharma V, Harafuji N, Belayew A, Chen YW. DUX4 differentially regulates transcriptomes of human rhabdomyosarcoma and mouse C2C12 cells. PLoS One 2013; 8:e64691. [PMID: 23717650 PMCID: PMC3661531 DOI: 10.1371/journal.pone.0064691] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 04/16/2013] [Indexed: 01/26/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is linked to the deletion of the D4Z4 arrays at chromosome 4q35. Recent studies suggested that aberrant expression of double homeobox 4 (DUX4) from the last D4Z4 repeat causes FSHD. The aim of this study is to determine transcriptomic responses to ectopically expressed DUX4 in human and mouse cells of muscle lineage. We expression profiled human rhabdomyosarcoma (RD) cells and mouse C2C12 cells transfected with expression vectors of DUX4 using the Affymetrix Human Genome U133 Plus 2.0 Arrays and Mouse Genome 430 2.0 Arrays, respectively. A total of 2267 and 150 transcripts were identified to be differentially expressed in the RD and C2C12 cells, respectively. Amongst the transcripts differentially expressed in the RD cells, MYOD and MYOG (2 fold, p<0.05), and six MYOD downstream targets were up-regulated in RD but not C2C12 cells. Furthermore, 13 transcripts involved in germline function were dramatically induced only in the RD cells expressing DUX4. The top 3 IPA canonical pathways affected by DUX4 were different between the RD (inflammation, BMP signaling and NRF-2 mediated oxidative stress) and the C2C12 cells (p53 signaling, cell cycle regulation and cellular energy metabolism). Amongst the 40 transcripts shared by the RD and C2C12 cells, UTS2 was significantly induced by 76 fold and 224 fold in the RD and C2C12 cells, respectively. The differential expression of MYOD, MYOG and UTS2 were validated using real-time quantitative RT-PCR. We further validated the differentially expressed genes in immortalized FSHD myoblasts and showed up-regulation of MYOD, MYOG, ZSCAN4 and UTS2. The results suggest that DUX4 regulates overlapped and distinct groups of genes and pathways in human and mouse cells as evident by the selective up-regulation of genes involved in myogenesis and gametogenesis in human RD and immortalized cells as well as the different molecular pathways identified in the cells.
Collapse
Affiliation(s)
- Vishakha Sharma
- Department of Molecular Medicine, George Washington University, Washington DC, United States of America
| | - Naoe Harafuji
- Center for Genetic Medicine Research, Children's National Medical Center, Washington DC, United States of America
| | | | - Yi-Wen Chen
- Center for Genetic Medicine Research, Children's National Medical Center, Washington DC, United States of America
- Department of Integrative Systems Biology, George Washington University, Washington DC, United States of America
| |
Collapse
|
80
|
Dmitriev P, Barat A, Polesskaya A, O'Connell MJ, Robert T, Dessen P, Walsh TA, Lazar V, Turki A, Carnac G, Laoudj-Chenivesse D, Lipinski M, Vassetzky YS. Simultaneous miRNA and mRNA transcriptome profiling of human myoblasts reveals a novel set of myogenic differentiation-associated miRNAs and their target genes. BMC Genomics 2013; 14:265. [PMID: 23597168 PMCID: PMC3639941 DOI: 10.1186/1471-2164-14-265] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Accepted: 03/26/2013] [Indexed: 01/10/2023] Open
Abstract
Background miRNA profiling performed in myogenic cells and biopsies from skeletal muscles has previously identified miRNAs involved in myogenesis. Results Here, we have performed miRNA transcriptome profiling in human affinity-purified CD56+ myoblasts induced to differentiate in vitro. In total, we have identified 60 miRNAs differentially expressed during myogenic differentiation. Many were not known for being differentially expressed during myogenic differentiation. Of these, 14 (miR-23b, miR-28, miR-98, miR-103, miR-107, miR-193a, miR-210, miR-324-5p, miR-324-3p, miR-331, miR-374, miR-432, miR-502, and miR-660) were upregulated and 6 (miR-31, miR-451, miR-452, miR-565, miR-594 and miR-659) were downregulated. mRNA transcriptome profiling performed in parallel resulted in identification of 6,616 genes differentially expressed during myogenic differentiation. Conclusions This simultaneous miRNA/mRNA transcriptome profiling allowed us to predict with high accuracy target genes of myogenesis-related microRNAs and to deduce their functions.
Collapse
Affiliation(s)
- Petr Dmitriev
- UMR 8126, Univ. Paris-Sud 11, CNRS, Institut de Cancérologie Gustave-Roussy, 39, rue Camille-Desmoulins, Villejuif 94805, France
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
81
|
Harafuji N, Schneiderat P, Walter MC, Chen YW. miR-411 is up-regulated in FSHD myoblasts and suppresses myogenic factors. Orphanet J Rare Dis 2013; 8:55. [PMID: 23561550 PMCID: PMC3637251 DOI: 10.1186/1750-1172-8-55] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Accepted: 03/22/2013] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant muscle disorder, which is linked to the contraction of the D4Z4 array at chromosome 4q35. Recent studies suggest that this shortening of the D4Z4 array leads to aberrant expression of double homeobox protein 4 (DUX4) and causes FSHD. In addition, misregulation of microRNAs (miRNAs) has been reported in muscular dystrophies including FSHD. In this study, we identified a miRNA that is differentially expressed in FSHD myoblasts and investigated its function. METHODS To identify misregulated miRNAs and their potential targets in FSHD myoblasts, we performed expression profiling of both miRNA and mRNA using TaqMan Human MicroRNA Arrays and Affymetrix Human Genome U133A plus 2.0 microarrays, respectively. In addition, we over-expressed miR-411 in C₂C₁₂ cells to determine the effect of miR-411 on myogenic markers. RESULTS Using miRNA and mRNA expression profiling, we identified 8 miRNAs and 1,502 transcripts that were differentially expressed in FSHD myoblasts during cell proliferation. One of the 8 differentially expressed miRNAs, miR-411, was validated by quantitative RT-PCR in both primary (2.1 fold, p<0.01) and immortalized (2.7 fold, p<0.01) myoblasts. In situ hybridization showed cytoplasmic localization of miR-411 in FSHD myoblasts. By analyzing both miRNA and mRNA data using Partek Genomics Suite, we identified 4 mRNAs potentially regulated by miR-411 including YY1 associated factor 2 (YAF2). The down-regulation of YAF2 in immortalized myoblasts was validated by immunoblotting (-3.7 fold, p<0.01). C₂C₁₂ cells were transfected with miR-411 to determine whether miR-411 affects YAF2 expression in myoblasts. The results showed that over-expression of miR-411 reduced YAF2 mRNA expression. In addition, expression of myogenic markers including Myod, myogenin, and myosin heavy chain 1 (Myh1) were suppressed by miR-411. CONCLUSIONS The study demonstrated that miR-411 was differentially expressed in FSHD myoblasts and may play a role in regulating myogenesis.
Collapse
Affiliation(s)
- Naoe Harafuji
- Center for Genetic Medicine Research, Children’s Research Institute, Washington, DC, USA
| | - Peter Schneiderat
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Maggie C Walter
- Friedrich-Baur-Institute, Department of Neurology, Ludwig-Maximilians-University of Munich, Munich, Germany
| | - Yi-Wen Chen
- Center for Genetic Medicine Research, Children’s Research Institute, Washington, DC, USA
- Department of Integrative Systems Biology and Department of Pediatrics, George Washington University, Washington, DC, USA
- Center for Genetic Medicine Research, Children’s National Medical Center, 111 Michigan Avenue, NW, Washington, DC 20010, USA
| |
Collapse
|
82
|
Pistoni M, Shiue L, Cline MS, Bortolanza S, Neguembor MV, Xynos A, Ares M, Gabellini D. Rbfox1 downregulation and altered calpain 3 splicing by FRG1 in a mouse model of Facioscapulohumeral muscular dystrophy (FSHD). PLoS Genet 2013; 9:e1003186. [PMID: 23300487 PMCID: PMC3536703 DOI: 10.1371/journal.pgen.1003186] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2011] [Accepted: 11/06/2012] [Indexed: 01/17/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a common muscle disease whose molecular pathogenesis remains largely unknown. Over-expression of FSHD region gene 1 (FRG1) in mice, frogs, and worms perturbs muscle development and causes FSHD-like phenotypes. FRG1 has been implicated in splicing, and we asked how splicing might be involved in FSHD by conducting a genome-wide analysis in FRG1 mice. We find that splicing perturbations parallel the responses of different muscles to FRG1 over-expression and disease progression. Interestingly, binding sites for the Rbfox family of splicing factors are over-represented in a subset of FRG1-affected splicing events. Rbfox1 knockdown, over-expression, and RNA-IP confirm that these are direct Rbfox1 targets. We find that FRG1 is associated to the Rbfox1 RNA and decreases its stability. Consistent with this, Rbfox1 expression is down-regulated in mice and cells over-expressing FRG1 as well as in FSHD patients. Among the genes affected is Calpain 3, which is mutated in limb girdle muscular dystrophy, a disease phenotypically similar to FSHD. In FRG1 mice and FSHD patients, the Calpain 3 isoform lacking exon 6 (Capn3 E6-) is increased. Finally, Rbfox1 knockdown and over-expression of Capn3 E6- inhibit muscle differentiation. Collectively, our results suggest that a component of FSHD pathogenesis may arise by over-expression of FRG1, reducing Rbfox1 levels and leading to aberrant expression of an altered Calpain 3 protein through dysregulated splicing.
Collapse
Affiliation(s)
- Mariaelena Pistoni
- Dulbecco Telethon Institute and Division of Regenerative Medicine, San Raffaele Scientific Institute, Milano, Italy
| | - Lily Shiue
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Melissa S. Cline
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Sergia Bortolanza
- Dulbecco Telethon Institute and Division of Regenerative Medicine, San Raffaele Scientific Institute, Milano, Italy
| | - Maria Victoria Neguembor
- Dulbecco Telethon Institute and Division of Regenerative Medicine, San Raffaele Scientific Institute, Milano, Italy
- Università Vita-Salute San Raffaele, Milano, Italy
| | - Alexandros Xynos
- Dulbecco Telethon Institute and Division of Regenerative Medicine, San Raffaele Scientific Institute, Milano, Italy
| | - Manuel Ares
- Department of Molecular, Cell, and Developmental Biology, University of California Santa Cruz, Santa Cruz, California, United States of America
| | - Davide Gabellini
- Dulbecco Telethon Institute and Division of Regenerative Medicine, San Raffaele Scientific Institute, Milano, Italy
| |
Collapse
|
83
|
Tassin A, Leroy B, Laoudj-Chenivesse D, Wauters A, Vanderplanck C, Le Bihan MC, Coppée F, Wattiez R, Belayew A. FSHD myotubes with different phenotypes exhibit distinct proteomes. PLoS One 2012; 7:e51865. [PMID: 23272181 PMCID: PMC3525578 DOI: 10.1371/journal.pone.0051865] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2012] [Accepted: 11/08/2012] [Indexed: 12/12/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a progressive muscle disorder linked to a contraction of the D4Z4 repeat array in the 4q35 subtelomeric region. This deletion induces epigenetic modifications that affect the expression of several genes located in the vicinity. In each D4Z4 element, we identified the double homeobox 4 (DUX4) gene. DUX4 expresses a transcription factor that plays a major role in the development of FSHD through the initiation of a large gene dysregulation cascade that causes myogenic differentiation defects, atrophy and reduced response to oxidative stress. Because miRNAs variably affect mRNA expression, proteomic approaches are required to define the dysregulated pathways in FSHD. In this study, we optimized a differential isotope protein labeling (ICPL) method combined with shotgun proteomic analysis using a gel-free system (2DLC-MS/MS) to study FSHD myotubes. Primary CD56(+) FSHD myoblasts were found to fuse into myotubes presenting various proportions of an atrophic or a disorganized phenotype. To better understand the FSHD myogenic defect, our improved proteomic procedure was used to compare predominantly atrophic or disorganized myotubes to the same matching healthy control. FSHD atrophic myotubes presented decreased structural and contractile muscle components. This phenotype suggests the occurrence of atrophy-associated proteolysis that likely results from the DUX4-mediated gene dysregulation cascade. The skeletal muscle myosin isoforms were decreased while non-muscle myosin complexes were more abundant. In FSHD disorganized myotubes, myosin isoforms were not reduced, and increased proteins were mostly involved in microtubule network organization and myofibrillar remodeling. A common feature of both FSHD myotube phenotypes was the disturbance of several caveolar proteins, such as PTRF and MURC. Taken together, our data suggest changes in trafficking and in the membrane microdomains of FSHD myotubes. Finally, the adjustment of a nuclear fractionation compatible with mass spectrometry allowed us to highlight alterations of proteins involved in mRNA processing and stability.
Collapse
Affiliation(s)
- Alexandra Tassin
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Baptiste Leroy
- Department of Proteomics and Microbiology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Dalila Laoudj-Chenivesse
- INSERM U1046 Physiologie et Médecine expérimentale Cœur et Muscle, CHU A. de Villeneuve, Montpellier, France
| | - Armelle Wauters
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Céline Vanderplanck
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Marie-Catherine Le Bihan
- University Pierre et Marie Curie- Paris 6, UM 76, INSERM U974, CNRS UMR 7215, Institut de Myologie, Paris, France
| | - Frédérique Coppée
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Ruddy Wattiez
- Department of Proteomics and Microbiology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | - Alexandra Belayew
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
- * E-mail:
| |
Collapse
|
84
|
Tassin A, Laoudj-Chenivesse D, Vanderplanck C, Barro M, Charron S, Ansseau E, Chen YW, Mercier J, Coppée F, Belayew A. DUX4 expression in FSHD muscle cells: how could such a rare protein cause a myopathy? J Cell Mol Med 2012. [PMID: 23206257 PMCID: PMC3823138 DOI: 10.1111/j.1582-4934.2012.01647.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is one of the most frequent hereditary muscle disorders. It is linked to contractions of the D4Z4 repeat array in 4q35. We have characterized the double homeobox 4 (DUX4) gene in D4Z4 and its mRNA transcribed from the distal D4Z4 unit to a polyadenylation signal in the flanking pLAM region. It encodes a transcription factor expressed in FSHD but not healthy muscle cells which initiates a gene deregulation cascade causing differentiation defects, muscle atrophy and oxidative stress. PITX1 was the first identified DUX4 target and encodes a transcription factor involved in muscle atrophy. DUX4 was found expressed in only 1/1000 FSHD myoblasts. We have now shown it was induced upon differentiation and detected in about 1/200 myotube nuclei. The DUX4 and PITX1 proteins presented staining gradients in consecutive myonuclei which suggested a diffusion as known for other muscle nuclear proteins. Both protein half-lifes were regulated by the ubiquitin-proteasome pathway. In addition, we could immunodetect the DUX4 protein in FSHD muscle extracts. As a model, we propose the DUX4 gene is stochastically activated in a small number of FSHD myonuclei. The resulting mRNAs are translated in the cytoplasm around an activated nucleus and the DUX4 proteins diffuse to adjacent nuclei where they activate target genes such as PITX1. The PITX1 protein can further diffuse to additional myonuclei and expand the transcriptional deregulation cascade initiated by DUX4. Together the diffusion and the deregulation cascade would explain how a rare protein could cause the muscle defects observed in FSHD.
Collapse
Affiliation(s)
- Alexandra Tassin
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Mons, Belgium
| | | | | | | | | | | | | | | | | | | |
Collapse
|
85
|
El Haddad M, Jean E, Turki A, Hugon G, Vernus B, Bonnieu A, Passerieux E, Hamade A, Mercier J, Laoudj-Chenivesse D, Carnac G. Glutathione peroxidase 3, a new retinoid target gene, is crucial for human skeletal muscle precursor cell survival. J Cell Sci 2012; 125:6147-56. [PMID: 23132926 DOI: 10.1242/jcs.115220] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Protection of satellite cells from cytotoxic damages is crucial to ensure efficient adult skeletal muscle regeneration and to improve therapeutic efficacy of cell transplantation in degenerative skeletal muscle diseases. It is therefore important to identify and characterize molecules and their target genes that control the viability of muscle stem cells. Recently, we demonstrated that high aldehyde dehydrogenase activity is associated with increased viability of human myoblasts. In addition to its detoxifying activity, aldehyde dehydrogenase can also catalyze the irreversible oxidation of vitamin A to retinoic acid; therefore, we examined whether retinoic acid is important for myoblast viability. We showed that when exposed to oxidative stress induced by hydrogen peroxide, adherent human myoblasts entered apoptosis and lost their capacity for adhesion. Pre-treatment with retinoic acid reduced the cytotoxic damage ex vivo and enhanced myoblast survival in transplantation assays. The effects of retinoic acid were maintained in dystrophic myoblasts derived from facioscapulohumeral patients. RT-qPCR analysis of antioxidant gene expression revealed glutathione peroxidase 3 (Gpx3), a gene encoding an antioxidant enzyme, as a potential retinoic acid target gene in human myoblasts. Knockdown of Gpx3 using short interfering RNA induced elevation in reactive oxygen species and cell death. The anti-cytotoxic effects of retinoic acid were impaired in GPx3-inactivated myoblasts, which indicates that GPx3 regulates the antioxidative effects of retinoic acid. Therefore, retinoid status and GPx3 levels may have important implications for the viability of human muscle stem cells.
Collapse
Affiliation(s)
- Marina El Haddad
- Inserm U1046, Université Montpellier 1, 34295 Montpellier, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
86
|
Turki A, Hayot M, Carnac G, Pillard F, Passerieux E, Bommart S, Raynaud de Mauverger E, Hugon G, Pincemail J, Pietri S, Lambert K, Belayew A, Vassetzky Y, Juntas Morales R, Mercier J, Laoudj-Chenivesse D. Functional muscle impairment in facioscapulohumeral muscular dystrophy is correlated with oxidative stress and mitochondrial dysfunction. Free Radic Biol Med 2012; 53:1068-79. [PMID: 22796148 DOI: 10.1016/j.freeradbiomed.2012.06.041] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Revised: 06/05/2012] [Accepted: 06/28/2012] [Indexed: 11/25/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD), the most frequent muscular dystrophy, is an autosomal dominant disease. In most individuals with FSHD, symptoms are restricted to muscles of the face, arms, legs, and trunk. FSHD is genetically linked to contractions of the D4Z4 repeat array causing activation of several genes. One of these maps in the repeat itself and expresses the DUX4 (the double homeobox 4) transcription factor causing a gene deregulation cascade. In addition, analyses of the RNA or protein expression profiles in muscle have indicated deregulations in the oxidative stress response. Since oxidative stress affects peripheral muscle function, we investigated mitochondrial function and oxidative stress in skeletal muscle biopsies and blood samples from patients with FSHD and age-matched healthy controls, and evaluated their association with physical performances. We show that specifically, oxidative stress (lipid peroxidation and protein carbonylation), oxidative damage (lipofuscin accumulation), and antioxidant enzymes (catalase, copper-zinc-dependent superoxide dismutase, and glutathione reductase) were higher in FSHD than in control muscles. FSHD muscles also presented abnormal mitochondrial function (decreased cytochrome c oxidase activity and reduced ATP synthesis). In addition, the ratio between reduced (GSH) and oxidized glutathione (GSSG) was strongly decreased in all FSHD blood samples as a consequence of GSSG accumulation. Patients with FSHD also had reduced systemic antioxidative response molecules, such as low levels of zinc (a SOD cofactor), selenium (a GPx cofactor involved in the elimination of lipid peroxides), and vitamin C. Half of them had a low ratio of gamma/alpha tocopherol and higher ferritin concentrations. Both systemic oxidative stress and mitochondrial dysfunction were correlated with functional muscle impairment. Mitochondrial ATP production was significantly correlated with both quadriceps endurance (T(LimQ)) and maximal voluntary contraction (MVC(Q)) values (rho=0.79, P=0.003; rho=0.62, P=0.05, respectively). The plasma concentration of oxidized glutathione was negatively correlated with the T(LimQ), MVC(Q) values, and the 2-min walk distance (MWT) values (rho=-0.60, P=0.03; rho=-0.56, P=0.04; rho=-0.93, P<0.0001, respectively). Our data characterized oxidative stress in patients with FSHD and demonstrated a correlation with their peripheral skeletal muscle dysfunction. They suggest that antioxidants that might modulate or delay oxidative insult may be useful in maintaining FSHD muscle functions.
Collapse
Affiliation(s)
- Ahmed Turki
- Université Montpellier 1 et Université Montpellier 2, INSERM, U1046, Montpellier, F-34000, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
87
|
Krom YD, Dumonceaux J, Mamchaoui K, den Hamer B, Mariot V, Negroni E, Geng LN, Martin N, Tawil R, Tapscott SJ, van Engelen BGM, Mouly V, Butler-Browne GS, van der Maarel SM. Generation of isogenic D4Z4 contracted and noncontracted immortal muscle cell clones from a mosaic patient: a cellular model for FSHD. THE AMERICAN JOURNAL OF PATHOLOGY 2012; 181:1387-401. [PMID: 22871573 DOI: 10.1016/j.ajpath.2012.07.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2011] [Revised: 06/25/2012] [Accepted: 07/02/2012] [Indexed: 01/24/2023]
Abstract
In most cases facioscapulohumeral muscular dystrophy (FSHD) is caused by contraction of the D4Z4 repeat in the 4q subtelomere. This contraction is associated with local chromatin decondensation and derepression of the DUX4 retrogene. Its complex genetic and epigenetic cause and high clinical variability in disease severity complicate investigations on the pathogenic mechanism underlying FSHD. A validated cellular model bypassing the considerable heterogeneity would facilitate mechanistic and therapeutic studies of FSHD. Taking advantage of the high incidence of somatic mosaicism for D4Z4 repeat contraction in de novo FSHD, we have established a clonal myogenic cell model from a mosaic patient. Individual clones are genetically identical except for the size of the D4Z4 repeat array, being either normal or FSHD sized. These clones retain their myogenic characteristics, and D4Z4 contracted clones differ from the noncontracted clones by the bursts of expression of DUX4 in sporadic nuclei, showing that this burst-like phenomenon is a locus-intrinsic feature. Consequently, downstream effects of DUX4 expression can be observed in D4Z4 contracted clones, like differential expression of DUX4 target genes. We also show their participation to in vivo regeneration with immunodeficient mice, further expanding the potential of these clones for mechanistic and therapeutic studies. These cell lines will facilitate pairwise comparisons to identify FSHD-specific differences and are expected to create new opportunities for high-throughput drug screens.
Collapse
Affiliation(s)
- Yvonne D Krom
- Leiden University Medical Center, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
88
|
Abstract
DUX4, a homeobox-containing gene present in a tandem array, is implicated in facioscapulohumeral muscular dystrophy (FSHD), a dominant autosomal disease. New findings about DUX4 have raised as many fundamental questions about the molecular pathology of this unique disease as they have answered. This review discusses recent studies addressing the question of whether there is extensive FSHD-related transcription dysregulation in adult-derived myoblasts and myotubes, the precursors for muscle repair. Two models for the role of DUX4 in FSHD are presented. One involves transient pathogenic expression of DUX4 in many cells in the muscle lineage before the myoblast stage resulting in a persistent, disease-related transcription profile ('Majority Rules'), which might be enhanced by subsequent oscillatory expression of DUX4. The other model emphasizes the toxic effects of inappropriate expression of DUX4 in only an extremely small percentage of FSHD myoblasts or myotube nuclei ('Minority Rules'). The currently favored Minority Rules model is not supported by recent studies of transcription dysregulation in FSHD myoblasts and myotubes. It also presents other difficulties, for example, explaining the expression of full-length DUX4 transcripts in FSHD fibroblasts. The Majority Rules model is the simpler explanation of findings about FSHD-associated gene expression and the DUX4-encoded homeodomain-type protein.
Collapse
|
89
|
A unique library of myogenic cells from facioscapulohumeral muscular dystrophy subjects and unaffected relatives: family, disease and cell function. Eur J Hum Genet 2011; 20:404-10. [PMID: 22108603 DOI: 10.1038/ejhg.2011.213] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
To explore possible mechanisms of pathology in facioscapulohumeral muscular dystrophy (FSHD), we generated a novel library of myogenic cells composed of paired cultures derived from FSHD subjects and unaffected first-degree relatives. We prepared cells from biopsies of both biceps and deltoid muscles obtained from each of 10 FSHD and 9 unaffected donors. We used this new collection to determine how family background and disease affected patterns of growth and differentiation, expression of a panel of candidate, and muscle-specific genes, and responses to exogenous stressors. We found that FSHD and unaffected cells had, on average, indistinguishable patterns of differentiation, gene expression, and dose-response curves to staurosporine, paraquat, hydrogen peroxide, and glutathione depletion. Differentiated FSHD and unaffected cultures were both more sensitive to glutathione depletion than proliferating cultures, but showed similar responses to paraquat, staurosporine, and peroxide. For stress responses, the sample size was sufficient to detect a 10% change in effect at the observed variability with a power of >99%. In contrast, for each of these properties, we found significant differences among cells from different cohorts, and these differences were independent of disease status, gender, or muscle biopsied. Thus, though none of the properties we examined could be used to reliably distinguish between FSHD and unaffected cells, family of origin was an important contributor to gene-expression patterns and stressor responses in cultures of both FSHD and unaffected myogenic cells.
Collapse
|
90
|
Pope BD, Tsumagari K, Battaglia D, Ryba T, Hiratani I, Ehrlich M, Gilbert DM. DNA replication timing is maintained genome-wide in primary human myoblasts independent of D4Z4 contraction in FSH muscular dystrophy. PLoS One 2011; 6:e27413. [PMID: 22096571 PMCID: PMC3214052 DOI: 10.1371/journal.pone.0027413] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 10/17/2011] [Indexed: 01/08/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is linked to contraction of an array of tandem 3.3-kb repeats (D4Z4) at 4q35.2 from 11-100 copies to 1-10 copies. The extent to which D4Z4 contraction at 4q35.2 affects overall 4q35.2 chromatin organization remains unclear. Because DNA replication timing is highly predictive of long-range chromatin interactions, we generated genome-wide replication-timing profiles for FSHD and control myogenic precursor cells. We compared non-immortalized myoblasts from four FSHD patients and three control individuals to each other and to a variety of other human cell types. This study also represents the first genome-wide comparison of replication timing profiles in non-immortalized human cell cultures. Myoblasts from both control and FSHD individuals all shared a myoblast-specific replication profile. In contrast, male and female individuals were readily distinguished by monoallelic differences in replication timing at DXZ4 and other regions across the X chromosome affected by X inactivation. We conclude that replication timing is a robust cell-type specific feature that is unaffected by FSHD-related D4Z4 contraction.
Collapse
Affiliation(s)
- Benjamin D. Pope
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Koji Tsumagari
- Human Genetics Program, Department of Biochemistry, and Tulane Cancer Center, Tulane Medical School, New Orleans, Louisiana, United States of America
| | - Dana Battaglia
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Tyrone Ryba
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Ichiro Hiratani
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| | - Melanie Ehrlich
- Human Genetics Program, Department of Biochemistry, and Tulane Cancer Center, Tulane Medical School, New Orleans, Louisiana, United States of America
| | - David M. Gilbert
- Department of Biological Science, Florida State University, Tallahassee, Florida, United States of America
| |
Collapse
|
91
|
Vanderplanck C, Ansseau E, Charron S, Stricwant N, Tassin A, Laoudj-Chenivesse D, Wilton SD, Coppée F, Belayew A. The FSHD atrophic myotube phenotype is caused by DUX4 expression. PLoS One 2011; 6:e26820. [PMID: 22053214 PMCID: PMC3203905 DOI: 10.1371/journal.pone.0026820] [Citation(s) in RCA: 144] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2011] [Accepted: 10/03/2011] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Facioscapulohumeral muscular dystrophy (FSHD) is linked to deletions in 4q35 within the D4Z4 repeat array in which we identified the double homeobox 4 (DUX4) gene. We found stable DUX4 mRNAs only derived from the most distal D4Z4 unit and unexpectedly extended to the flanking pLAM region that provided an intron and a polyadenylation signal. DUX4 encodes a transcription factor expressed in FSHD but not control primary myoblasts or muscle biopsies. The DUX4 protein initiates a large transcription deregulation cascade leading to muscle atrophy and oxidative stress, which are FSHD key features. METHODOLOGY/PRINCIPAL FINDINGS We now show that transfection of myoblasts with a DUX4 expression vector leads to atrophic myotube formation associated with the induction of E3 ubiquitin ligases (MuRF1 and Atrogin1/MAFbx) typical of muscle atrophy. DUX4 induces expression of downstream targets deregulated in FSHD such as mu-crystallin and TP53. We developed specific siRNAs and antisense oligonucleotides (AOs) targeting the DUX4 mRNA. Addition of these antisense agents to primary FSHD myoblast cultures suppressed DUX4 protein expression and affected expression of the above-mentioned markers. CONCLUSIONS/SIGNIFICANCE These results constitute a proof of concept for the development of therapeutic approaches for FSHD targeting DUX4 expression.
Collapse
MESH Headings
- Animals
- Biomarkers/metabolism
- Cells, Cultured
- Down-Regulation/drug effects
- Gene Expression Regulation/drug effects
- Homeodomain Proteins/genetics
- Homeodomain Proteins/metabolism
- Humans
- Mice
- Models, Biological
- Muscle Fibers, Skeletal/drug effects
- Muscle Fibers, Skeletal/metabolism
- Muscle Fibers, Skeletal/pathology
- Muscle Proteins/metabolism
- Muscular Atrophy/metabolism
- Muscular Atrophy/pathology
- Muscular Dystrophy, Facioscapulohumeral/metabolism
- Muscular Dystrophy, Facioscapulohumeral/pathology
- Oligonucleotides, Antisense/pharmacology
- Phenotype
- RNA Interference/drug effects
- RNA Splicing/drug effects
- RNA Splicing/genetics
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Small Interfering/metabolism
- SKP Cullin F-Box Protein Ligases/metabolism
- Transfection
- Tripartite Motif Proteins
- Ubiquitin-Protein Ligases/metabolism
Collapse
Affiliation(s)
| | - Eugénie Ansseau
- Laboratory of Molecular Biology, University of Mons, Mons, Belgium
| | | | - Nadia Stricwant
- Laboratory of Molecular Biology, University of Mons, Mons, Belgium
| | - Alexandra Tassin
- Laboratory of Molecular Biology, University of Mons, Mons, Belgium
| | | | - Steve D. Wilton
- Molecular Genetic Therapy Group, University of Western Australia, Nedlands, Australia
| | | | | |
Collapse
|
92
|
Richards M, Coppée F, Thomas N, Belayew A, Upadhyaya M. Facioscapulohumeral muscular dystrophy (FSHD): an enigma unravelled? Hum Genet 2011; 131:325-40. [PMID: 21984394 DOI: 10.1007/s00439-011-1100-z] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 09/26/2011] [Indexed: 01/02/2023]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is the third most common muscular dystrophy after the dystrophinopathies and myotonic dystrophy and is associated with a typical pattern of muscle weakness. Most patients with FSHD carry a large deletion in the polymorphic D4Z4 macrosatellite repeat array at 4q35 and present with 1-10 repeats whereas non-affected individuals possess 11-150 repeats. An almost identical repeat array is present at 10q26 and the high sequence identity between these two arrays can cause difficulties in molecular diagnosis. Each 3.3-kb D4Z4 unit contains a DUX4 (double homeobox 4) gene that, among others, is activated upon contraction of the 4q35 repeat array due to the induction of chromatin remodelling of the 4qter region. A number of 4q subtelomeric sequence variants are now recognised, although FSHD only occurs in association with three 'permissive' haplotypes, each of which is associated with a polyadenylation signal located immediately distal of the last D4Z4 unit. The resulting poly-A tail appears to stabilise DUX4 mRNAs transcribed from this most distal D4Z4 unit in FSHD muscle cells. Synthesis of both the DUX4 transcripts and protein in FSHD muscle cells induces significant cell toxicity. DUX4 is a transcription factor that may target several genes which results in a deregulation cascade which inhibits myogenesis, sensitises cells to oxidative stress and induces muscle atrophy, thus recapitulating many of the key molecular features of FSHD.
Collapse
Affiliation(s)
- Mark Richards
- School of Medicine, Institute of Medical Genetics, Cardiff University, Cardiff, CF14 4XN, UK
| | | | | | | | | |
Collapse
|
93
|
Tsumagari K, Chang SC, Lacey M, Baribault C, Chittur SV, Sowden J, Tawil R, Crawford GE, Ehrlich M. Gene expression during normal and FSHD myogenesis. BMC Med Genomics 2011; 4:67. [PMID: 21951698 PMCID: PMC3204225 DOI: 10.1186/1755-8794-4-67] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2011] [Accepted: 09/27/2011] [Indexed: 01/31/2023] Open
Abstract
Background Facioscapulohumeral muscular dystrophy (FSHD) is a dominant disease linked to contraction of an array of tandem 3.3-kb repeats (D4Z4) at 4q35. Within each repeat unit is a gene, DUX4, that can encode a protein containing two homeodomains. A DUX4 transcript derived from the last repeat unit in a contracted array is associated with pathogenesis but it is unclear how. Methods Using exon-based microarrays, the expression profiles of myogenic precursor cells were determined. Both undifferentiated myoblasts and myoblasts differentiated to myotubes derived from FSHD patients and controls were studied after immunocytochemical verification of the quality of the cultures. To further our understanding of FSHD and normal myogenesis, the expression profiles obtained were compared to those of 19 non-muscle cell types analyzed by identical methods. Results Many of the ~17,000 examined genes were differentially expressed (> 2-fold, p < 0.01) in control myoblasts or myotubes vs. non-muscle cells (2185 and 3006, respectively) or in FSHD vs. control myoblasts or myotubes (295 and 797, respectively). Surprisingly, despite the morphologically normal differentiation of FSHD myoblasts to myotubes, most of the disease-related dysregulation was seen as dampening of normal myogenesis-specific expression changes, including in genes for muscle structure, mitochondrial function, stress responses, and signal transduction. Other classes of genes, including those encoding extracellular matrix or pro-inflammatory proteins, were upregulated in FSHD myogenic cells independent of an inverse myogenesis association. Importantly, the disease-linked DUX4 RNA isoform was detected by RT-PCR in FSHD myoblast and myotube preparations only at extremely low levels. Unique insights into myogenesis-specific gene expression were also obtained. For example, all four Argonaute genes involved in RNA-silencing were significantly upregulated during normal (but not FSHD) myogenesis relative to non-muscle cell types. Conclusions DUX4's pathogenic effect in FSHD may occur transiently at or before the stage of myoblast formation to establish a cascade of gene dysregulation. This contrasts with the current emphasis on toxic effects of experimentally upregulated DUX4 expression at the myoblast or myotube stages. Our model could explain why DUX4's inappropriate expression was barely detectable in myoblasts and myotubes but nonetheless linked to FSHD.
Collapse
Affiliation(s)
- Koji Tsumagari
- Human Genetics Program, Tulane Medical School, New Orleans, LA, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Dmitriev P, Petrov A, Ansseau E, Stankevicins L, Charron S, Kim E, Bos TJ, Robert T, Turki A, Coppée F, Belayew A, Lazar V, Carnac G, Laoudj D, Lipinski M, Vassetzky YS. The Krüppel-like factor 15 as a molecular link between myogenic factors and a chromosome 4q transcriptional enhancer implicated in facioscapulohumeral dystrophy. J Biol Chem 2011; 286:44620-31. [PMID: 21937448 DOI: 10.1074/jbc.m111.254052] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD), a dominant hereditary disease with a prevalence of 7 per 100,000 individuals, is associated with a partial deletion in the subtelomeric D4Z4 repeat array on chromosome 4q. The D4Z4 repeat contains a strong transcriptional enhancer that activates promoters of several FSHD-related genes. We report here that the enhancer within the D4Z4 repeat binds the Krüppel-like factor KLF15. KLF15 was found to be up-regulated during myogenic differentiation induced by serum starvation or by overexpression of the myogenic differentiation factor MYOD. When overexpressed, KLF15 activated the D4Z4 enhancer and led to overexpression of DUX4c (Double homeobox 4, centromeric) and FRG2 (FSHD region gene 2) genes, whereas its silencing caused inactivation of the D4Z4 enhancer. In immortalized human myoblasts, the D4Z4 enhancer was activated by the myogenic factor MYOD, an effect that was abolished upon KLF15 silencing or when the KLF15-binding sites within the D4Z4 enhancer were mutated, indicating that the myogenesis-related activation of the D4Z4 enhancer was mediated by KLF15. KLF15 and several myogenesis-related factors were found to be expressed at higher levels in myoblasts, myotubes, and muscle biopsies from FSHD patients than in healthy controls. We propose that KLF15 serves as a molecular link between myogenic factors and the activity of the D4Z4 enhancer, and it thus contributes to the overexpression of the DUX4c and FRG2 genes during normal myogenic differentiation and in FSHD.
Collapse
Affiliation(s)
- Petr Dmitriev
- CNRS UMR8126, Université Paris-Sud 11, Institut de Cancérologie Gustave Roussy, 94805 Villejuif, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Jean E, Laoudj-Chenivesse D, Notarnicola C, Rouger K, Serratrice N, Bonnieu A, Gay S, Bacou F, Duret C, Carnac G. Aldehyde dehydrogenase activity promotes survival of human muscle precursor cells. J Cell Mol Med 2011; 15:119-33. [PMID: 19840193 PMCID: PMC3822499 DOI: 10.1111/j.1582-4934.2009.00942.x] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Aldehyde dehydrogenases (ALDH) are a family of enzymes that efficiently detoxify aldehydic products generated by reactive oxygen species and might therefore participate in cell survival. Because ALDH activity has been used to identify normal and malignant cells with stem cell properties, we asked whether human myogenic precursor cells (myoblasts) could be identified and isolated based on their levels of ALDH activity. Human muscle explant-derived cells were incubated with ALDEFLUOR, a fluorescent substrate for ALDH, and we determined by flow cytometry the level of enzyme activity. We found that ALDH activity positively correlated with the myoblast-CD56+ fraction in those cells, but, we also observed heterogeneity of ALDH activity levels within CD56-purified myoblasts. Using lentiviral mediated expression of shRNA we demonstrated that ALDH activity was associated with expression of Aldh1a1 protein. Surprisingly, ALDH activity and Aldh1a1 expression levels were very low in mouse, rat, rabbit and non-human primate myoblasts. Using different approaches, from pharmacological inhibition of ALDH activity by diethylaminobenzaldehyde, an inhibitor of class I ALDH, to cell fractionation by flow cytometry using the ALDEFLUOR assay, we characterized human myoblasts expressing low or high levels of ALDH. We correlated high ALDH activity ex vivo to resistance to hydrogen peroxide (H2O2)-induced cytotoxic effect and in vivo to improved cell viability when human myoblasts were transplanted into host muscle of immune deficient scid mice. Therefore detection of ALDH activity, as a purification strategy, could allow non-toxic and efficient isolation of a fraction of human myoblasts resistant to cytotoxic damage.
Collapse
Affiliation(s)
- Elise Jean
- INSERM, ERI 25, Muscle et Pathologies, Montpellier, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
96
|
Cheli S, François S, Bodega B, Ferrari F, Tenedini E, Roncaglia E, Ferrari S, Ginelli E, Meneveri R. Expression profiling of FSHD-1 and FSHD-2 cells during myogenic differentiation evidences common and distinctive gene dysregulation patterns. PLoS One 2011; 6:e20966. [PMID: 21695143 PMCID: PMC3113851 DOI: 10.1371/journal.pone.0020966] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2010] [Accepted: 05/17/2011] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Determine global gene dysregulation affecting 4q-linked (FSHD-1) and non 4q-linked (FSHD-2) cells during early stages of myogenic differentiation. This approach has been never applied to FSHD pathogenesis. METHODOLOGY/PRINCIPAL FINDINGS By in vitro differentiation of FSHD-1 and FSHD-2 myoblasts and gene chip analysis we derived that gene expression profile is altered only in FSHD-1 myoblasts and FSHD-2 myotubes. The changes seen in FSHD-1 regarded a general defect in cell cycle progression, probably due to the upregulation of myogenic markers PAX3 and MYOD1, and a deficit of factors (SUV39H1 and HMGB2) involved in D4Z4 chromatin conformation. On the other hand, FSHD-2 mytubes were characterized by a general defect in RNA metabolism, protein synthesis and degradation and, to a lesser extent, in cell cycle. Common dysregulations regarded genes involved in response to oxidative stress and in sterol biosynthetic process. Interestingly, our results also suggest that miRNAs might be implied in both FSHD-1 and FSHD-2 gene dysregulation. Finally, in both cell differentiation systems, we did not observe a gradient of altered gene expression throughout the 4q35 chromosome. CONCLUSIONS/SIGNIFICANCE FSHD-1 and FSHD-2 cells showed, in different steps of myogenic differentiation, a global deregulation of gene expression rather than an alteration of expression of 4q35 specific genes. In general, FSHD-1 and FSHD-2 global gene deregulation interested common and distinctive biological processes. In this regard, defects of cell cycle progression (FSHD-1 and to a lesser extent FSHD-2), protein synthesis and degradation (FSHD-2), response to oxidative stress (FSHD-1 and FSHD-2), and cholesterol homeostasis (FSHD-1 and FSHD-2) may in general impair a correct myogenesis. Taken together our results recapitulate previously reported defects of FSHD-1, and add new insights into the gene deregulation characterizing both FSHD-1 and FSHD-2, in which miRNAs may play a role.
Collapse
Affiliation(s)
- Stefania Cheli
- Department of Biology and Genetics for Medical Sciences, University of Milan, Milan, Italy
| | - Stephanie François
- Department of Experimental Medicine, University of Milano-Bicocca, Monza, Italy
| | - Beatrice Bodega
- Department of Biology and Genetics for Medical Sciences, University of Milan, Milan, Italy
| | - Francesco Ferrari
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Elena Tenedini
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrica Roncaglia
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Sergio Ferrari
- Department of Biomedical Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Enrico Ginelli
- Department of Biology and Genetics for Medical Sciences, University of Milan, Milan, Italy
| | - Raffaella Meneveri
- Department of Experimental Medicine, University of Milano-Bicocca, Monza, Italy
| |
Collapse
|
97
|
Decreased proliferation kinetics of mouse myoblasts overexpressing FRG1. PLoS One 2011; 6:e19780. [PMID: 21603621 PMCID: PMC3095625 DOI: 10.1371/journal.pone.0019780] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2011] [Accepted: 04/04/2011] [Indexed: 11/24/2022] Open
Abstract
Although recent publications have linked the molecular events driving facioscapulohumeral muscular dystrophy (FSHD) to expression of the double homeobox transcription factor DUX4, overexpression of FRG1 has been proposed as one alternative causal agent as mice overexpressing FRG1 present with muscular dystrophy. Here, we characterize proliferative defects in two independent myoblast lines overexpressing FRG1. Myoblasts isolated from thigh muscle of FRG1 transgenic mice, an affected dystrophic muscle, exhibit delayed proliferation as measured by decreased clone size, whereas myoblasts isolated from the unaffected diaphragm muscle proliferated normally. To confirm the observation that overexpression of FRG1 could impair myoblast proliferation, we examined C2C12 myoblasts with inducible overexpression of FRG1, finding increased doubling time and G1-phase cells in mass culture after induction of FRG1 and decreased levels of pRb phosphorylation. We propose that depressed myoblast proliferation may contribute to the pathology of mice overexpressing FRG1 and may play a part in FSHD.
Collapse
|
98
|
Morgan JE, Zammit PS. Direct effects of the pathogenic mutation on satellite cell function in muscular dystrophy. Exp Cell Res 2010; 316:3100-8. [DOI: 10.1016/j.yexcr.2010.05.014] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 05/03/2010] [Indexed: 12/14/2022]
|
99
|
Wuebbles RD, Long SW, Hanel ML, Jones PL. Testing the effects of FSHD candidate gene expression in vertebrate muscle development. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2010; 3:386-400. [PMID: 20490329 PMCID: PMC2872745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 03/02/2010] [Accepted: 03/23/2010] [Indexed: 05/29/2023]
Abstract
The genetic lesion leading to facioscapulohumeral muscular dystrophy (FSHD) is a dominant deletion at the 4q35 locus. The generally accepted disease model involves an epigenetic dysregulation in the region resulting in the upregulation of one or more proximal genes whose overexpression specifically affects skeletal muscle. However, multiple FSHD candidate genes have been proposed without clear consensus. Using Xenopus laevis as a model for vertebrate development our lab has studied the effects of overexpression of the FSHD candidate gene ortholog, frg1 (FSHD region gene 1), showing that increased levels of frg1 systemically led specifically to an abnormal musculature and increased angiogenesis, the two most prominent clinical features of FSHD. Here we studied the overexpression effects of three other promising FSHD candidate genes, DUX4, DUX4c, and PITX1 using the same model system and methods for direct comparison. Expression of even very low levels of either DUX4 or pitx1 early in development led to massive cellular loss and severely abnormal development. These abnormalities were not muscle specific. In contrast, elevated levels of DUX4c resulted in no detectable adverse affects on muscle and DUX4c levels did not alter the expression of myogenic regulators. This data supports a model for DUX4 and PITX1 in FSHD only as pro-apoptotic factors if their expression in FSHD is confined to cells within the myogenic pathway; neither could account for the vascular pathology prevalent in FSHD. Taken together, increased frg1 expression alone leads to a phenotype that most closely resembles the pathophysiology observed in FSHD patients.
Collapse
Affiliation(s)
- Ryan D Wuebbles
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, B107 Chemical and Life Sciences Laboratory, Urbana, IL 61801, USA
| | | | | | | |
Collapse
|
100
|
Klooster R, Straasheijm K, Shah B, Sowden J, Frants R, Thornton C, Tawil R, van der Maarel S. Comprehensive expression analysis of FSHD candidate genes at the mRNA and protein level. Eur J Hum Genet 2009; 17:1615-24. [PMID: 19809486 PMCID: PMC2987013 DOI: 10.1038/ejhg.2009.62] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2008] [Revised: 03/18/2009] [Accepted: 03/18/2009] [Indexed: 01/24/2023] Open
Abstract
In facioscapulohumeral muscular dystrophy (FSHD) the majority of patients carry a D4Z4 macrosatellite repeat contraction in the subtelomere of chromosome 4q. Several disease mechanisms have been proposed to explain how repeat contraction causes muscular dystrophy. All proposed mechanisms foresee a change from a closed to a more open chromatin structure followed by loss of control over expression of genes in or proximal to D4Z4. Initially, a distance and residual repeat size-dependent upregulation of the candidate genes FRG2, FRG1 and ANT1 was observed, but most successive expression studies failed to support transcriptional upregulation of 4qter genes. Moreover, chromatin studies do not provide evidence for a cis-spreading mechanism operating at 4qter in FSHD. In part, this inconsistency may be explained by differences in the techniques used, and the use of RNA samples obtained from different muscle groups. The aim of this study is to comprehensively and uniformly study the expression of the FSHD candidate genes FRG1, FRG2, CRYM, ANT1, ALP, PITX1 and LRP2BP at the RNA and protein level in identically processed primary myoblasts, myotubes and quadriceps muscle. Expression was compared between samples obtained from FSHD patients and normal controls with samples from myotonic dystrophy type 1 patients as disease controls. No consistent changes in RNA or protein expression levels were observed between the samples. The one exception was a selective increase in FRG2 mRNA expression in FSHD myotubes. This study provides further evidence that there is no demonstrable consistent, large magnitude, overexpression of any of the FSHD candidate genes.
Collapse
Affiliation(s)
- Rinse Klooster
- Department of Human Genetics, Leiden University Medical Center, Leiden, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|