1
|
Crisafulli O, Baptista R, Drid P, Grattarola L, Bottoni G, Lavaselli E, Negro M, Tupler R, Quintiero V, D'Antona G. Analysis of Body Fluid Distribution, Phase Angle and Its Association With Maximal Oxygen Consumption in Facioscapulohumeral Dystrophy: An Observational Study. Health Sci Rep 2025; 8:e70335. [PMID: 39807483 PMCID: PMC11726644 DOI: 10.1002/hsr2.70335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/20/2024] [Accepted: 12/29/2024] [Indexed: 01/16/2025] Open
Abstract
Background and Aims Body composition parameters associated with aerobic fitness, mirrored by maximal oxygen consumption (V̇O2max), have recently gained interest as indicators of physical efficiency in facioscapulohumeral dystrophy (FSHD). Bioimpedance analysis (BIA) allows a noninvasive and repeatable estimate of body composition but is based on the use of predictive equations which, if used in cohorts with different characteristics from those for which the equation was originally formulated, could give biased results. Instead, the phase angle (PhA), a BIA raw bioelectrical parameter reflecting body fluids distribution, could provide reliable data for such analysis. Methods 33 clinically and genetically characterized FSHD patients (mean age 35.7; 10 females) and 27 sex and age-matched healthy controls (HC) were included in the analysis. BIA was used to evaluate body fluids distribution (intracellular water [ICW], extracellular water [ECW], and total body water [TBW]), and PhA, while cardiopulmonary exercise test was used to estimate V̇O2max. Results The groups were comparable for ECW and TBW. Instead, patients showed lower values of ICW (p = 0.020), ICW/ECW ratio (p < 0.001), and PhA (p < 0.001). Moreover, patients reported lower V̇O2max (p = 0.001 for absolute values; p = 0.002 for values expressed in relation to body weight) which, unlike HC, was not associated to PhA. Conclusion Based on our results, PhA of FSHD patients is lower than HC. Since PhA mirrors the ICW/ECW ratio, the lower share of ICW seems to be the basis of such difference. Given the lack of association with V̇O2max, PhA cannot be considered a reliable indicator of aerobic fitness in FSHD.
Collapse
Affiliation(s)
- Oscar Crisafulli
- CRIAMS‐Sport Medicine Centre VogheraUniversity of PaviaVogheraItaly
- Faculty of Sport and Physical EducationUniversity of Novi SadNovi SadSerbia
| | - Renato Baptista
- Department of Research and DevelopmentLUNEXDifferdangeLuxembourg
- Luxembourg Health & Sport Sciences Research Institute A.s.b.l.DifferdangeLuxembourg
| | - Patrik Drid
- Faculty of Sport and Physical EducationUniversity of Novi SadNovi SadSerbia
| | - Luca Grattarola
- CRIAMS‐Sport Medicine Centre VogheraUniversity of PaviaVogheraItaly
| | - Giorgio Bottoni
- CRIAMS‐Sport Medicine Centre VogheraUniversity of PaviaVogheraItaly
| | | | - Massimo Negro
- CRIAMS‐Sport Medicine Centre VogheraUniversity of PaviaVogheraItaly
| | - Rossella Tupler
- Department of Life SciencesUniversity of Modena and Reggio EmiliaModenaItaly
| | - Venere Quintiero
- CRIAMS‐Sport Medicine Centre VogheraUniversity of PaviaVogheraItaly
| | - Giuseppe D'Antona
- CRIAMS‐Sport Medicine Centre VogheraUniversity of PaviaVogheraItaly
- Department of Public Health, Experimental and Forensic MedicineUniversity of PaviaPaviaItaly
| |
Collapse
|
2
|
Hitti G, Kavanaugh A, Zukotynski B, Billi F. Management of FSHD symptoms: current assistive technologies and pharmacological approaches. Disabil Rehabil Assist Technol 2024:1-9. [PMID: 39565698 DOI: 10.1080/17483107.2024.2431058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Facioscapulohumeral Muscular Dystrophy (FSHD) is a genetically linked disorder characterized by the progressive deterioration of muscles controlling facial and scapular movement. The severity and distribution of affected muscle groups vary significantly across patient demographics, necessitating diverse assistive approaches. OBJECTIVE This review aims to evaluate the effectiveness of assistive devices and therapeutic options, including medications and rehabilitative therapies, tailored to specific manifestations of FSHD. METHODS An analysis of existing literature and clinical trials was conducted to explore current assistive technologies and pharmacological treatments, focusing on their application to FSHD patients with varying symptom severity. RESULTS Although several pharmacological treatments, such as steroids, supplements, protein inhibitors, and knockout strategies, are under investigation, none have yet fully counteracted the disease process driven by toxic DUX4 production. Consequently, the broad assortment of assistive devices currently on the market remain critical for improving quality of life. CONCLUSION Despite advances in pharmacological research, the variability in FSHD manifestations necessitates a personalized approach combining assistive technologies and tailored therapeutic interventions. Future research should continue exploring integrative strategies to address the unique needs of FSHD patients.
Collapse
Affiliation(s)
- George Hitti
- UCLA Orthopedic Surgery Research Center, Los Angeles, USA
| | | | | | - Fabrizio Billi
- UCLA Orthopedic Surgery Research Center, Los Angeles, USA
| |
Collapse
|
3
|
Moriggi M, Ruggiero L, Torretta E, Zoppi D, Arosio B, Ferri E, Castegna A, Fiorillo C, Gelfi C, Capitanio D. Muscle Proteome Analysis of Facioscapulohumeral Dystrophy Patients Reveals a Metabolic Rewiring Promoting Oxidative/Reductive Stress Contributing to the Loss of Muscle Function. Antioxidants (Basel) 2024; 13:1406. [PMID: 39594549 PMCID: PMC11591206 DOI: 10.3390/antiox13111406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is caused by the epigenetic de-repression of the double homeobox 4 (DUX4) gene, leading to asymmetric muscle weakness and atrophy that begins in the facial and scapular muscles and progresses to the lower limbs. This incurable condition can severely impair muscle function, ultimately resulting in a loss of ambulation. A thorough analysis of molecular factors associated with the varying degrees of muscle impairment in FSHD is still lacking. This study investigates the molecular mechanisms and biomarkers in the biceps brachii of FSHD patients, classified according to the FSHD clinical score, the A-B-C-D classification scheme, and global proteomic variation. Our findings reveal distinct metabolic signatures and compensatory responses in patients. In severe cases, we observe pronounced metabolic dysfunction, marked by dysregulated glycolysis, activation of the reductive pentose phosphate pathway (PPP), a shift toward a reductive TCA cycle, suppression of oxidative phosphorylation, and an overproduction of antioxidants that is not matched by an increase in the redox cofactors needed for their function. This imbalance culminates in reductive stress, exacerbating muscle wasting and inflammation. In contrast, mild cases show metabolic adaptations that mitigate stress by activating polyols and the oxidative PPP, preserving partial energy flow through the oxidative TCA cycle, which supports mitochondrial function and energy balance. Furthermore, activation of the hexosamine biosynthetic pathway promotes autophagy, protecting muscle cells from apoptosis. In conclusion, our proteomic data indicate that specific metabolic alterations characterize both mild and severe FSHD patients. Molecules identified in mild cases may represent potential diagnostic and therapeutic targets for FSHD.
Collapse
Affiliation(s)
- Manuela Moriggi
- Department of Biomedical Sciences for Health, University of Milan, Via Luigi Mangiagalli 31, 20133 Milan, Italy; (M.M.); (D.C.)
| | - Lucia Ruggiero
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, Via Sergio Pansini 5, 80131 Naples, Italy; (L.R.); (D.Z.)
| | - Enrica Torretta
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy;
| | - Dario Zoppi
- Department of Neurosciences, Reproductive and Odontostomatological Sciences, University of Naples “Federico II”, Via Sergio Pansini 5, 80131 Naples, Italy; (L.R.); (D.Z.)
| | - Beatrice Arosio
- Department of Clinical Sciences and Community Health, University of Milan, Via della Commenda 19, 20122 Milan, Italy;
| | - Evelyn Ferri
- IRCCS Ca’ Granda Ospedale Maggiore Policlinico Foundation, Via Francesco Sforza 35, 20122 Milan, Italy;
| | - Alessandra Castegna
- Department of Biosciences, Biotechnologies and Environment, University of Bari ALDO MORO, Via Orabona 4, 70125 Bari, Italy;
| | - Chiara Fiorillo
- Child Neuropsychiatric Unit, IRCCS Istituto Giannina Gaslini, DINOGMI-University of Genova, Via Gerolamo Gaslini 5, 16147 Genova, Italy;
| | - Cecilia Gelfi
- Department of Biomedical Sciences for Health, University of Milan, Via Luigi Mangiagalli 31, 20133 Milan, Italy; (M.M.); (D.C.)
- Laboratory of Proteomics and Lipidomics, IRCCS Orthopedic Institute Galeazzi, Via R. Galeazzi 4, 20161 Milan, Italy;
| | - Daniele Capitanio
- Department of Biomedical Sciences for Health, University of Milan, Via Luigi Mangiagalli 31, 20133 Milan, Italy; (M.M.); (D.C.)
| |
Collapse
|
4
|
Attarian S, Beloribi-Djefaflia S, Bernard R, Nguyen K, Cances C, Gavazza C, Echaniz-Laguna A, Espil C, Evangelista T, Feasson L, Audic F, Zagorda B, Milhe De Bovis V, Stojkovic T, Sole G, Salort-Campana E, Sacconi S. French National Protocol for diagnosis and care of facioscapulohumeral muscular dystrophy (FSHD). J Neurol 2024; 271:5778-5803. [PMID: 38955828 DOI: 10.1007/s00415-024-12538-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 06/21/2024] [Accepted: 06/23/2024] [Indexed: 07/04/2024]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is one of the most common genetically inherited myopathies in adults. It is characterized by incomplete penetrance and variable expressivity. Typically, FSHD patients display asymmetric weakness of facial, scapular, and humeral muscles that may progress to other muscle groups, particularly the abdominal and lower limb muscles. Early-onset patients display more severe muscle weakness and atrophy, resulting in a higher frequency of associated skeletal abnormalities. In these patients, multisystem involvement, including respiratory, ocular, and auditory, is more frequent and severe and may include the central nervous system. Adult-onset FSHD patients may also display some degree of multisystem involvement which mainly remains subclinical. In 95% of cases, FSHD patients carry a pathogenic contraction of the D4Z4 repeat units (RUs) in the subtelomeric region of chromosome 4 (4q35), which leads to the expression of DUX4 retrogene, toxic for muscles (FSHD1). Five percent of patients display the same clinical phenotype in association with a mutation in the SMCHD1 gene located in chromosome 18, inducing epigenetic modifications of the 4q D4Z4 repeated region and expression of DUX4 retrogene. This review highlights the complexities and challenges of diagnosing and managing FSHD, underscoring the importance of standardized approaches for optimal patient outcomes. It emphasizes the critical role of multidisciplinary care in addressing the diverse manifestations of FSHD across different age groups, from skeletal abnormalities in early-onset cases to the often-subclinical multisystem involvement in adults. With no current cure, the focus on alleviating symptoms and slowing disease progression through coordinated care is paramount.
Collapse
Affiliation(s)
- Shahram Attarian
- Reference Center for Neuromuscular Disorders and ALS, Timone University Hospital, Aix-Marseille University, Marseille, France.
- FILNEMUS, European Reference Network for Rare Diseases (ERN-NMD), Marseille, France.
- Marseille Medical Genetics, Aix Marseille Université-Inserm UMR_1251, 13005, Marseille, France.
| | - Sadia Beloribi-Djefaflia
- Reference Center for Neuromuscular Disorders and ALS, Timone University Hospital, Aix-Marseille University, Marseille, France
| | - Rafaelle Bernard
- Marseille Medical Genetics, Aix Marseille Université-Inserm UMR_1251, 13005, Marseille, France
| | - Karine Nguyen
- Marseille Medical Genetics, Aix Marseille Université-Inserm UMR_1251, 13005, Marseille, France
| | - Claude Cances
- Reference Center for Neuromuscular Disorders, Toulouse Children's Hospital, Toulouse, France
- Pediatric Neurology Department, Toulouse Children's Hospital, Toulouse, France
| | - Carole Gavazza
- Reference Center for Neuromuscular Disorders and ALS, Timone University Hospital, Aix-Marseille University, Marseille, France
| | - Andoni Echaniz-Laguna
- Department of Neurology, APHP, CHU de Bicêtre, Le Kremlin Bicêtre, France
- French National Reference Center for Rare Neuropathies (NNERF), Le Kremlin Bicêtre, France
- Inserm U1195, University Paris Saclay, Le Kremlin Bicêtre, France
| | - Caroline Espil
- Reference Center for Neuromuscular Disorders AOC, Children's Hospital, CHU Bordeaux, Bordeaux, France
| | - Teresinha Evangelista
- Institute of Myology, Nord/Est/Ile-de-France Neuromuscular Reference Center, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| | - Léonard Feasson
- Department of Clinical and Exercise Physiology, University Hospital Center of Saint-Etienne, 42000, Saint-Etienne, France
- Inter-University Laboratory of Human Movement Biology, EA 7424, Jean Monnet University, 42000, Saint-Etienne, France
| | - Frédérique Audic
- Reference Center for Neuromuscular Diseases in Children PACARARE, Neuropediatrics Department, Timone University Children's Hospital, Marseille, France
| | - Berenice Zagorda
- Department of Clinical and Exercise Physiology, University Hospital Center of Saint-Etienne, 42000, Saint-Etienne, France
| | - Virginie Milhe De Bovis
- Reference Center for Neuromuscular Disorders and ALS, Timone University Hospital, Aix-Marseille University, Marseille, France
| | - Tanya Stojkovic
- Institute of Myology, Nord/Est/Ile-de-France Neuromuscular Reference Center, Pitié-Salpêtrière Hospital, APHP, Sorbonne University, Paris, France
| | - Guilhem Sole
- Centre de Référence des Maladies Neuromusculaires AOC, FILNEMUS, Hôpital Pellegrin, CHU de Bordeaux, Bordeaux, France
| | - Emmanuelle Salort-Campana
- Reference Center for Neuromuscular Disorders and ALS, Timone University Hospital, Aix-Marseille University, Marseille, France
| | - Sabrina Sacconi
- Peripheral Nervous System and Muscle Department, Université Côte d'Azur, CHU Nice, Pasteur 2, Nice Hospital, France.
| |
Collapse
|
5
|
Wilson VD, Bommart S, Passerieux E, Thomas C, Pincemail J, Picot MC, Mercier J, Portet F, Arbogast S, Laoudj-Chenivesse D. Muscle strength, quantity and quality and muscle fat quantity and their association with oxidative stress in patients with facioscapulohumeral muscular dystrophy: Effect of antioxidant supplementation. Free Radic Biol Med 2024; 219:112-126. [PMID: 38574978 DOI: 10.1016/j.freeradbiomed.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/19/2024] [Accepted: 04/01/2024] [Indexed: 04/06/2024]
Abstract
The purpose of this study was to identify causes of quadriceps muscle weakness in facioscapulohumeral muscular dystrophy (FSHD). To this aim, we evaluated quadriceps muscle and fat volumes by magnetic resonance imaging and their relationships with muscle strength and oxidative stress markers in adult patients with FSHD (n = 32) and healthy controls (n = 7), and the effect of antioxidant supplementation in 20 of the 32 patients with FSHD (n = 10 supplementation and n = 10 placebo) (NCT01596803). Compared with healthy controls, the dominant quadriceps strength and quality (muscle strength per unit of muscle volume) were decreased in patients with FSHD. In addition, fat volume was increased, without changes in total muscle volume. Moreover, in patients with FSHD, the lower strength of the non-dominant quadriceps was associated with lower muscle quality compared with the dominant muscle. Antioxidant supplementation significantly changed muscle and fat volumes in the non-dominant quadriceps, and muscle quality in the dominant quadriceps. This was associated with improved muscle strength (both quadriceps) and antioxidant response. These findings suggest that quadriceps muscle strength decline may not be simply explained by atrophy and may be influenced also by the muscle intrinsic characteristics. As FSHD is associated with increased oxidative stress, supplementation might reduce oxidative stress and increase antioxidant defenses, promoting changes in muscle function.
Collapse
Affiliation(s)
- Vinicius Dias Wilson
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; Centro Universitário Estácio de Belo Horizonte, Minas Gerais, Brazil.
| | - Sébastien Bommart
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; Department of Radiology, CHU of Montpellier, Arnaud de Villeneuve Hospital, 34090, Montpellier, France.
| | - Emilie Passerieux
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France.
| | - Claire Thomas
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; LBEPS, Univ Evry, IRBA, University Paris Saclay, 91025, Evry, France.
| | - Joël Pincemail
- Department of CREDEC, Department of Medical Chemistry, University Hospital of Liege, Sart Tilman, Liege, Belgium.
| | - Marie Christine Picot
- Department of Biostatistics and Epidemiology, University Hospital, Montpellier, France; CIC 1001-INSERM, Montpellier, France.
| | - Jacques Mercier
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; Department of Clinical Physiology, CHU of Montpellier, Montpellier, France.
| | - Florence Portet
- Department of Clinical Physiology, CHU of Montpellier, Montpellier, France; U1061 INSERM, CHU de Montpellier, Montpellier University, France.
| | - Sandrine Arbogast
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France.
| | - Dalila Laoudj-Chenivesse
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; Department of Clinical Physiology, CHU of Montpellier, Montpellier, France.
| |
Collapse
|
6
|
Cedillo-Servin G, Dahri O, Meneses J, van Duijn J, Moon H, Sage F, Silva J, Pereira A, Magalhães FD, Malda J, Geijsen N, Pinto AM, Castilho M. 3D Printed Magneto-Active Microfiber Scaffolds for Remote Stimulation and Guided Organization of 3D In Vitro Skeletal Muscle Models. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307178. [PMID: 37950402 DOI: 10.1002/smll.202307178] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/11/2023] [Indexed: 11/12/2023]
Abstract
This work reports the rational design and fabrication of magneto-active microfiber meshes with controlled hexagonal microstructures via melt electrowriting (MEW) of a magnetized polycaprolactone-based composite. In situ iron oxide nanoparticle deposition on oxidized graphene yields homogeneously dispersed magnetic particles with sizes above 0.5 µm and low aspect ratio, preventing cellular internalization and toxicity. With these fillers, homogeneous magnetic composites with high magnetic content (up to 20 weight %) are obtained and processed in a solvent-free manner for the first time. MEW of magnetic composites enabled the creation of skeletal muscle-inspired design of hexagonal scaffolds with tunable fiber diameter, reconfigurable modularity, and zonal distribution of magneto-active and nonactive material, with elastic tensile deformability. External magnetic fields below 300 mT are sufficient to trigger out-of-plane reversible deformation. In vitro culture of C2C12 myoblasts on three-dimensional (3D) Matrigel/collagen/MEW scaffolds showed that microfibers guided the formation of 3D myotube architectures, and the presence of magnetic particles does not significantly affect viability or differentiation rates after 8 days. Centimeter-sized skeletal muscle constructs allowed for reversible, continued, and dynamic magneto-mechanical stimulation. Overall, these innovative microfiber scaffolds provide magnetically deformable platforms suitable for dynamic culture of skeletal muscle, offering potential for in vitro disease modeling.
Collapse
Affiliation(s)
- Gerardo Cedillo-Servin
- Department of Orthopaedics, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, 3508 GA, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, 5612 AE, The Netherlands
| | - Ouafa Dahri
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, 2333 ZC, The Netherlands
- Leiden Node, The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden, 2333 ZA, The Netherlands
| | - João Meneses
- Departamento de Engenharia Química, Faculty of Engineering, University of Porto, Porto, 4200-465, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, 4200-465, Portugal
| | - Joost van Duijn
- Department of Orthopaedics, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, 3508 GA, The Netherlands
| | - Harrison Moon
- Department of Orthopaedics, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, 3508 GA, The Netherlands
| | - Fanny Sage
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, 2333 ZC, The Netherlands
- Leiden Node, The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden, 2333 ZA, The Netherlands
| | - Joana Silva
- Departamento de Engenharia Química, Faculty of Engineering, University of Porto, Porto, 4200-465, Portugal
| | - André Pereira
- Departamento de Engenharia Química, Faculty of Engineering, University of Porto, Porto, 4200-465, Portugal
| | - Fernão D Magalhães
- Departamento de Engenharia Química, Faculty of Engineering, University of Porto, Porto, 4200-465, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, 4200-465, Portugal
| | - Jos Malda
- Department of Orthopaedics, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, 3508 GA, The Netherlands
- Department of Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, 3508 GA, The Netherlands
| | - Niels Geijsen
- Department of Anatomy and Embryology, Leiden University Medical Center, Leiden, 2333 ZC, The Netherlands
- Leiden Node, The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden, 2333 ZA, The Netherlands
| | - Artur M Pinto
- Departamento de Engenharia Química, Faculty of Engineering, University of Porto, Porto, 4200-465, Portugal
- ALiCE - Associate Laboratory in Chemical Engineering, Faculty of Engineering, University of Porto, Porto, 4200-465, Portugal
| | - Miguel Castilho
- Department of Orthopaedics, Regenerative Medicine Center Utrecht, University Medical Center Utrecht, Utrecht, 3508 GA, The Netherlands
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, 5612 AE, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, 5612 AE, The Netherlands
| |
Collapse
|
7
|
Murray R, Donnelly CM, Drescher KD, Graham CD. The lived experience of facioscapulohumeral muscular dystrophy: A systematic review and synthesis of the qualitative literature. Muscle Nerve 2024; 69:7-17. [PMID: 37691606 DOI: 10.1002/mus.27964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Revised: 08/17/2023] [Accepted: 08/20/2023] [Indexed: 09/12/2023]
Abstract
INTRODUCTION In this review we sought to characterize the lived experience of people living with FSHD (pwFSHD) to help clinicians to orient their services to the needs of these individuals. METHODS Five electronic databases were systematically searched for qualitative research studies containing quotations from pwFSHD. ENhancing Transparency in REporting the Synthesis of Qualitative research and Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines informed the methodology. Study quality was assessed using the Critical Appraisal Skills Programme Checklist tool, which measures the methodological quality of qualitative research. Data extracted from included studies were analyzed using thematic synthesis. RESULTS Ninety-nine pwFSHD took part in the six studies included in this review - from research teams based in two countries. Five descriptive themes emerged: "Engaging with life as symptoms progress"; "The emotional journey"; "A family burden to bear"; "Social connection and disconnection"; and "Tension between visibility and invisibility." From these, two analytical themes were derived: "The emotional challenge of continuing and intensifying adaptation" and "The relational burden of rare disease." DISCUSSION The lived experience of pwFSHD is characterized by physical, emotional, and social challenges that impact on engagement with life, particularly as symptoms progress. Further research is needed to provide a fuller understanding of the experience of pain in FSHD and of the lived experience of FSHD across cultures.
Collapse
Affiliation(s)
- Roisin Murray
- Psychology Service for Older People, Western Health and Social Care Trust, Londonderry, UK
| | - Clare M Donnelly
- Psychological Therapies Service, Northern Health and Social Care Trust, Antrim, UK
| | - Kent D Drescher
- National Center for PTSD, VA Palo Alto Healthcare System, Palo Alto, California, USA
| | - Christopher D Graham
- Psychology Service for Older People, Western Health and Social Care Trust, Londonderry, UK
- School of Psychological Sciences and Health, University of Strathclyde, Glasgow, UK
| |
Collapse
|
8
|
Amzali S, Wilson VD, Bommart S, Picot MC, Galas S, Mercier J, Poucheret P, Cristol JP, Arbogast S, Laoudj-Chenivesse D. Nutritional Status of Patients with Facioscapulohumeral Muscular Dystrophy. Nutrients 2023; 15:nu15071673. [PMID: 37049513 PMCID: PMC10096775 DOI: 10.3390/nu15071673] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 04/14/2023] Open
Abstract
In patients with facioscapulohumeral muscular dystrophy (FSHD), a rare genetic neuromuscular disease, reduced physical performance is associated with lower blood levels of vitamin C, zinc, selenium, and increased oxidative stress markers. Supplementation of vitamin C, vitamin E, zinc, and selenium improves the quadriceps' physical performance. Here, we compared the nutritional status of 74 women and 85 men with FSHD. Calorie intake was lower in women with FSHD than in men. Moreover, we assessed vitamin C, vitamin E, zinc, copper, and selenium intakes in diet and their concentrations in the plasma. Vitamin E, copper, and zinc intake were lower in women with FSHD than in men, whereas plasma vitamin C, copper levels, and copper/zinc ratio were higher in women with FSHD than in men. The dietary intake and plasma concentrations of the studied vitamins and minerals were not correlated in both sexes. A well-balanced and varied diet might not be enough in patients with FSHD to correct the observed vitamin/mineral deficiencies. A low energy intake is a risk factor for suboptimal intake of proteins, vitamins, and minerals that are important for protein synthesis and other metabolic pathways and that might contribute to progressive muscle mass loss. Antioxidant supplementation and higher protein intake seem necessary to confer protection against oxidative stress and skeletal muscle mass loss.
Collapse
Affiliation(s)
- Sedda Amzali
- PhyMedExp, Université de Montpellier, INSERM, CNRS, CHU de Montpellier, 34295 Montpellier, France
| | - Vinicius Dias Wilson
- Departamento de Educação Física, Centro Universitário Estácio de Belo Horizonte, Belo Horizonte 30411-052, Minas Gerais, Brazil
- Pró-Reitoria de Assuntos Comunitários e Estudantis, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina 39100-000, Minas Gerais, Brazil
| | - Sébastien Bommart
- PhyMedExp, Université de Montpellier, INSERM, CNRS, CHU de Montpellier, 34295 Montpellier, France
- Service de Radiologie, Hôpital Arnaud-de-Villeneuve, CHU de Montpellier, 34295 Montpellier, France
| | - Marie-Christine Picot
- Clinical Research and Epidemiology Unit (Department of Medicale Information), Centre d'Investigation Clinique 1411 INSERM, CHU Montpellier, Univ Montpellier, CEDEX 5, 34295 Montpellier, France
| | - Simon Galas
- Institut des Biomolecules Max Mousseron (IBMM), Centre National de Recherche Scientifique (CNRS), University of Montpellier, ENSCM, 34000 Montpellier, France
| | - Jacques Mercier
- PhyMedExp, Université de Montpellier, INSERM, CNRS, CHU de Montpellier, 34295 Montpellier, France
- Department of Clinical Physiology, CHU of Montpellier, 34295 Montpellier, France
| | - Patrick Poucheret
- Qualisud, Université de Montpellier, CIRAD, Institut Agro, IRD, Avignon Université, Université de La Réunion, 34000 Montpellier, France
| | - Jean-Paul Cristol
- PhyMedExp, Université de Montpellier, INSERM, CNRS, CHU de Montpellier, 34295 Montpellier, France
- Department of Biochemistry, University Hospital of Montpellier, 34295 Montpellier, France
| | - Sandrine Arbogast
- PhyMedExp, Université de Montpellier, INSERM, CNRS, CHU de Montpellier, 34295 Montpellier, France
| | - Dalila Laoudj-Chenivesse
- PhyMedExp, Université de Montpellier, INSERM, CNRS, CHU de Montpellier, 34295 Montpellier, France
- Department of Clinical Physiology, CHU of Montpellier, 34295 Montpellier, France
| |
Collapse
|
9
|
Muacevic A, Adler JR. Anticipation Avoids Adversity: Anesthetic Management of a Case of Facioscapulohumeral Dystrophy (FSHD). Cureus 2023; 15:e34442. [PMID: 36733549 PMCID: PMC9888316 DOI: 10.7759/cureus.34442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 02/01/2023] Open
Abstract
Patients with muscular dystrophies, especially those pauci-symptomatic presenting for surgery pose a complex problem for the anesthesiologist in preparing, optimizing and performing anesthesia. A myriad of complications including cardiac, respiratory, rhabdomyolysis, hyperkalemia, increased sensitivity to muscle relaxants etc., influence the anesthetic technique and recovery. Preoperative identification and appropriate choice of anesthesia technique can prevent most of the adverse events during anesthesia. We present a case of facioscapulohumeral dystrophy (FSHD) presenting for emergency appendectomy. Preoperative investigations and lung function were adequate. The patient underwent general anesthesia with propofol, cisatarcurium, and remifentanil and was maintained on total IV anesthesia for the duration of surgery. Continuous neuromuscular monitoring was carried out at two sites and the patient responded normally to intubating dose of cisatracurium and subsequent top-up doses, showing no increased sensitivity or need for dose reduction. The patient was hemodynamically stable with propofol and remifentanil infusions, with inhalational agents purposefully avoided. The patient was reversed with anticholinesterase and good train-of-four (TOF) ratio (>90%) was ensured before being shifted to recovery. The patient had a further uneventful course in the hospital.
Collapse
|
10
|
ANT1 overexpression models: Some similarities with facioscapulohumeral muscular dystrophy. Redox Biol 2022; 56:102450. [PMID: 36030628 PMCID: PMC9434167 DOI: 10.1016/j.redox.2022.102450] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/04/2022] [Accepted: 08/17/2022] [Indexed: 11/20/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disorder characterized by progressive muscle weakness. Adenine nucleotide translocator 1 (ANT1), the only 4q35 gene involved in mitochondrial function, is strongly expressed in FSHD skeletal muscle biopsies. However, its role in FSHD is unclear. In this study, we evaluated ANT1 overexpression effects in primary myoblasts from healthy controls and during Xenopus laevis organogenesis. We also compared ANT1 overexpression effects with the phenotype of FSHD muscle cells and biopsies. Here, we report that the ANT1 overexpression-induced phenotype presents some similarities with FSHD muscle cells and biopsies. ANT1-overexpressing muscle cells showed disorganized morphology, altered cytoskeletal arrangement, enhanced mitochondrial respiration/glycolysis, ROS production, oxidative stress, mitochondrial fragmentation and ultrastructure alteration, as observed in FSHD muscle cells. ANT1 overexpression in Xenopus laevis embryos affected skeletal muscle development, impaired skeletal muscle, altered mitochondrial ultrastructure and led to oxidative stress as observed in FSHD muscle biopsies. Moreover, ANT1 overexpression in X. laevis embryos affected heart structure and mitochondrial ultrastructure leading to cardiac arrhythmia, as described in some patients with FSHD. Overall our data suggest that ANT1 could contribute to mitochondria dysfunction and oxidative stress in FSHD muscle cells by modifying their bioenergetic profile associated with ROS production. Such interplay between energy metabolism and ROS production in FSHD will be of significant interest for future prospects.
Collapse
|
11
|
Heher P, Ganassi M, Weidinger A, Engquist EN, Pruller J, Nguyen TH, Tassin A, Declèves AE, Mamchaoui K, Banerji CRS, Grillari J, Kozlov AV, Zammit PS. Interplay between mitochondrial reactive oxygen species, oxidative stress and hypoxic adaptation in facioscapulohumeral muscular dystrophy: Metabolic stress as potential therapeutic target. Redox Biol 2022; 51:102251. [PMID: 35248827 PMCID: PMC8899416 DOI: 10.1016/j.redox.2022.102251] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 01/25/2022] [Indexed: 12/13/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is characterised by descending skeletal muscle weakness and wasting. FSHD is caused by mis-expression of the transcription factor DUX4, which is linked to oxidative stress, a condition especially detrimental to skeletal muscle with its high metabolic activity and energy demands. Oxidative damage characterises FSHD and recent work suggests metabolic dysfunction and perturbed hypoxia signalling as novel pathomechanisms. However, redox biology of FSHD remains poorly understood, and integrating the complex dynamics of DUX4-induced metabolic changes is lacking. Here we pinpoint the kinetic involvement of altered mitochondrial ROS metabolism and impaired mitochondrial function in aetiology of oxidative stress in FSHD. Transcriptomic analysis in FSHD muscle biopsies reveals strong enrichment for pathways involved in mitochondrial complex I assembly, nitrogen metabolism, oxidative stress response and hypoxia signalling. We found elevated mitochondrial ROS (mitoROS) levels correlate with increases in steady-state mitochondrial membrane potential in FSHD myogenic cells. DUX4 triggers mitochondrial membrane polarisation prior to oxidative stress generation and apoptosis through mitoROS, and affects mitochondrial health through lipid peroxidation. We identify complex I as the primary target for DUX4-induced mitochondrial dysfunction, with strong correlation between complex I-linked respiration and cellular oxygenation/hypoxia signalling activity in environmental hypoxia. Thus, FSHD myogenesis is uniquely susceptible to hypoxia-induced oxidative stress as a consequence of metabolic mis-adaptation. Importantly, mitochondria-targeted antioxidants rescue FSHD pathology more effectively than conventional antioxidants, highlighting the central involvement of disturbed mitochondrial ROS metabolism. This work provides a pathomechanistic model by which DUX4-induced changes in oxidative metabolism impair muscle function in FSHD, amplified when metabolic adaptation to varying O2 tension is required.
Collapse
Affiliation(s)
- Philipp Heher
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK.
| | - Massimo Ganassi
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - Adelheid Weidinger
- Ludwig Boltzmann Institute for Traumatology. The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Elise N Engquist
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - Johanna Pruller
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - Thuy Hang Nguyen
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000, Mons, Belgium
| | - Alexandra Tassin
- Laboratory of Respiratory Physiology, Pathophysiology and Rehabilitation, Research Institute for Health Sciences and Technology, University of Mons, 7000, Mons, Belgium
| | - Anne-Emilie Declèves
- Department of Metabolic and Molecular Biochemistry, Research Institute for Health Sciences and Technology, University of Mons, 7000, Mons, Belgium
| | - Kamel Mamchaoui
- Institut de Myologie, Sorbonne University, INSERM UMRS974, Paris, France
| | - Christopher R S Banerji
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK
| | - Johannes Grillari
- Ludwig Boltzmann Institute for Traumatology. The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria; Institute for Molecular Biotechnology, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Andrey V Kozlov
- Ludwig Boltzmann Institute for Traumatology. The Research Center in Cooperation with AUVA, Vienna, Austria; Austrian Cluster for Tissue Regeneration, Vienna, Austria
| | - Peter S Zammit
- Randall Centre for Cell and Molecular Biophysics, King's College London, Guy's Campus, London, UK.
| |
Collapse
|
12
|
Jia FF, Drew AP, Nicholson GA, Corbett A, Kumar KR. Facioscapulohumeral muscular dystrophy type 2: an update on the clinical, genetic, and molecular findings. Neuromuscul Disord 2021; 31:1101-1112. [PMID: 34711481 DOI: 10.1016/j.nmd.2021.09.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 09/01/2021] [Accepted: 09/09/2021] [Indexed: 11/25/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a common genetic disease of the skeletal muscle with a characteristic pattern of weakness. Facioscapulohumeral muscular dystrophy type 2 (FSHD2) accounts for approximately 5% of all cases of FSHD and describes patients without a D4Z4 repeat contraction on chromosome 4. Phenotypically FSHD2 shows virtually no difference from FSHD1 and both forms of FSHD arise via a common downstream mechanism of epigenetic derepression of the transcription factor DUX4 in skeletal muscle cells. This results in expression of DUX4 and target genes leading to skeletal muscle toxicity. Over the past decade, major progress has been made in our understanding of the genetic and epigenetic architecture that underlies FSHD2 pathogenesis, as well as the clinical manifestations and disease progression. These include the finding that FSHD2 is a digenic disease and that mutations in the genes SMCHD1, DNMT3B, and more recently LRIF1, can cause FSHD2. FSHD2 is complex and it is important that clinicians keep abreast of recent developments; this review aims to serve as an update of the clinical, genetic, and molecular research into this condition.
Collapse
Affiliation(s)
- Fangzhi Frank Jia
- Department of Neurology, Concord Repatriation General Hospital, Concord, New South Wales 2139, Australia.
| | - Alexander P Drew
- Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia.
| | - Garth Alexander Nicholson
- Department of Neurology, Concord Repatriation General Hospital, Concord, New South Wales 2139, Australia; Molecular Medicine Laboratory, Concord Repatriation General Hospital, Concord, New South Wales 2139, Australia; Northcott Neuroscience Laboratory, ANZAC Research Institute, Concord, New South Wales 2139, Australia; Sydney Medical School, University of Sydney, Camperdown, New South Wales 2050, Australia.
| | - Alastair Corbett
- Department of Neurology, Concord Repatriation General Hospital, Concord, New South Wales 2139, Australia; Sydney Medical School, University of Sydney, Camperdown, New South Wales 2050, Australia.
| | - Kishore Raj Kumar
- Department of Neurology, Concord Repatriation General Hospital, Concord, New South Wales 2139, Australia; Kinghorn Centre for Clinical Genomics, Garvan Institute of Medical Research, Darlinghurst, New South Wales 2010, Australia; Molecular Medicine Laboratory, Concord Repatriation General Hospital, Concord, New South Wales 2139, Australia; Sydney Medical School, University of Sydney, Camperdown, New South Wales 2050, Australia.
| |
Collapse
|
13
|
Ng CY, Wu F. Scapular winging secondary to serratus anterior dysfunction: analysis of clinical presentations and etiology in a consecutive series of 96 patients. J Shoulder Elbow Surg 2021; 30:2336-2343. [PMID: 33675974 DOI: 10.1016/j.jse.2021.02.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 02/03/2021] [Accepted: 02/08/2021] [Indexed: 02/01/2023]
Abstract
BACKGROUND This study aimed to establish the relative incidence of etiologies causing serratus anterior (SA) dysfunction in patients with proven abnormality on needle electromyography. METHODS This was a retrospective review of patients with scapular winging secondary to SA dysfunction. Each patient underwent a detailed clinical, radiological, and neurophysiological assessment to arrive at the precise etiological diagnosis. Patients with atypical clinical features were referred for a neurologist's assessment. Hematological and genetic testing were requested at the discretion of the neurologist. A scapular winging severity score based on clinical signs was devised to aid clinical grading. RESULTS Between 2014 and 2020, a consecutive series of 108 patients with suspected SA dysfunction were assessed, of whom 96 met the inclusion criteria. There were 34 females and 62 males, with a mean age of 38 years (range, 15-77 years). Winging affected the right scapulae in 69 patients, the left scapulae in 17 patients, and was bilateral in 10 patients. This was caused by a myopathic disorder in 12 (12%) patients. Eighty-four (88%) patients had a long thoracic nerve lesion, caused by cervical pathology (2), iatrogenic injury (2), trauma (33), and neuralgic amyotrophy (NA) (47). Among those with NA, winging resolved spontaneously within 3 years of onset in 22 patients (mean duration, 16 months; range, 3-36 months). No patients recovered fully if their duration of winging lasted longer than 3 years. Patients with palsy secondary to NA tended to have a worse severity of winging than those due to a traumatic cause (P = .04). CONCLUSION NA accounted for approximately half of the patients with SA dysfunction; therefore, it is essential to also consider the differentials of myopathy, trauma, iatrogenic injury, and spinal pathology. We recommend the judicious employment of ancillary tests and a low threshold of referral to a neurologist, in order to arrive at the exact diagnosis to accurately guide patient treatment.
Collapse
Affiliation(s)
- Chye Yew Ng
- Upper Limb Unit, Wrightington Hospital, Appley Bridge, Wigan, UK.
| | - Feiran Wu
- Department of Orthopaedics, University Hospitals Birmingham, Birmingham, UK
| |
Collapse
|
14
|
CRISPR mediated targeting of DUX4 distal regulatory element represses DUX4 target genes dysregulated in Facioscapulohumeral muscular dystrophy. Sci Rep 2021; 11:12598. [PMID: 34131248 PMCID: PMC8206090 DOI: 10.1038/s41598-021-92096-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 06/04/2021] [Indexed: 11/08/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a debilitating muscle disease that currently does not have an effective cure or therapy. The abnormal reactivation of DUX4, an embryonic gene that is epigenetically silenced in somatic tissues, is causal to FSHD. Disease-specific reactivation of DUX4 has two common characteristics, the presence of a non-canonical polyadenylation sequence within exon 3 of DUX4 that stabilizes pathogenic transcripts, and the loss of repressive chromatin modifications at D4Z4, the macrosatellite repeat which encodes DUX4. We used CRISPR/Cas9 to silence DUX4 using two independent approaches. We deleted the DUX4 pathogenic polyadenylation signal, which resulted in downregulation of pathogenic DUX4-fl transcripts. In another approach, we transcriptionally repressed DUX4 by seeding heterochromatin using the dCas9-KRAB platform within exon 3. These feasibility of targeting DUX4 experiments were initially tested in a non-myogenic carcinoma cell line that we have previously characterized. Subsequently, in an immortalized patient myoblast cell line, we demonstrated that targeting DUX4 by either approach led to substantial downregulation of not only pathogenic DUX4 transcripts, but also a subset of its target genes that are known biomarkers of FSHD. These findings offer proof-of-concept of the effect of silencing the polyadenylation sequence on pathogenic DUX4 expression.
Collapse
|
15
|
Rehn JA, O’Connor MJ, White DL, Yeung DT. DUX Hunting-Clinical Features and Diagnostic Challenges Associated with DUX4-Rearranged Leukaemia. Cancers (Basel) 2020; 12:cancers12102815. [PMID: 33007870 PMCID: PMC7599557 DOI: 10.3390/cancers12102815] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2020] [Revised: 09/26/2020] [Accepted: 09/29/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary DUX4-rearrangement (DUX4r) is a recently discovered recurrent genomic lesion reported in 4–7% of childhood B cell acute lymphoblastic leukaemia (B-ALL) cases. This subtype has favourable outcomes, especially in children and adolescents treated with intensive chemotherapy. The fusion most commonly links the hypervariable IGH gene to DUX4 a gene located within the D4Z4 macrosatellite repeat on chromosome 4. DUX4r is cryptic to most standard diagnostic techniques, and difficult to identify even with next generation sequencing assays. This review summarises the clinical features and molecular genetics of DUX4r B-ALL and proposes prospective new diagnostic methods. Abstract DUX4-rearrangement (DUX4r) is a recently discovered recurrent genomic lesion reported in 4–7% of childhood B cell acute lymphoblastic leukaemia (B-ALL) cases. This subtype has favourable outcomes, especially in children and adolescents treated with intensive chemotherapy. The fusion most commonly links the hypervariable IGH gene to DUX4 a gene located within the D4Z4 macrosatellite repeat on chromosome 4, with a homologous polymorphic repeat on chromosome 10. DUX4r is cryptic to most standard diagnostic techniques, and difficult to identify even with next generation sequencing assays. This review summarises the clinical features and molecular genetics of DUX4r B-ALL and proposes prospective new diagnostic methods.
Collapse
Affiliation(s)
- Jacqueline A. Rehn
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (J.A.R.); (D.T.Y.)
- Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA 5000, Australia
| | - Matthew J. O’Connor
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (J.A.R.); (D.T.Y.)
- Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA 5000, Australia
- Michael Rice Centre for Haematology and Oncology, Womens’s & Children’s Hospital, North Adelaide, SA 5006, Australia
| | - Deborah L. White
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (J.A.R.); (D.T.Y.)
- Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA 5000, Australia
- Australian Genomics, The Murdoch Children’s Research Institute, Parkville, VIC 3052, Australia
- Australian and New Zealand Children’s Oncology Group (ANZCHOG), Clayton, VIC 3168, Australia
- Correspondence:
| | - David T. Yeung
- Cancer Program, Precision Medicine Theme, South Australian Health & Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia; (J.A.R.); (D.T.Y.)
- Faculty of Health and Medical Science, University of Adelaide, Adelaide, SA 5000, Australia
- Department of Haematology, Royal Adelaide Hospital, Adelaide, SA 5000, Australia
| |
Collapse
|
16
|
[Pain management in rare diseases]. Schmerz 2020; 34:447-459. [PMID: 32734404 DOI: 10.1007/s00482-020-00487-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
In this article we address the relevance of rare diseases and their peculiarities with respect to pain therapy. Towards this end, four rare diseases (hemophilia, Morbus Fabry, dermatomyositis, and facioscapulohumeral dystrophy (FSHD)) will be presented and fundamental aspects of their pain therapies described. The diseases were chosen to showcase a pain therapy based on the WHO-step-by-step plan (hemophilia), a complex but established pain therapy (M. Fabry), and two less well established, individually adapted pain therapies (dermatomyositis, FSHD).
Collapse
|
17
|
Karauzum S, Hangul C, Bozkurt S, Bilge U, Ozdem S, Altunbas H, Uysal H, Koc F. The ratios of estradiol and progesterone to testosterone influence the severity of facioscapulohumeral muscular dystrophy. NEUROL SCI NEUROPHYS 2020. [DOI: 10.4103/nsn.nsn_37_20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
18
|
Jacques MF, Stockley RC, Onambele-Pearson GL, Reeves ND, Stebbings GK, Dawson EA, Groves L, Morse CI. Quality of life in adults with muscular dystrophy. Health Qual Life Outcomes 2019; 17:121. [PMID: 31307472 PMCID: PMC6632211 DOI: 10.1186/s12955-019-1177-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2018] [Accepted: 06/06/2019] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Muscle weakness is a defining characteristic of Muscular Dystrophy (MD); however, yet while speculated, objective measures of muscle weakness has not been reported in relation to quality of life in adults with MD. OBJECTIVES 1) compare the self-reported QoL of adults with Duchenne MD (DMD), Beckers MD (BMD), Limb-Girdle MD (LGMD) and Fascioscapulohumeral MD (FSHD, and a non-MD (CTRL) group; 2) present and compare between groups measures of Impairment (Muscle Strength and Activities of Daily Living) and Perception (Fatigue, Pain and Self-Efficacy); and 3) identify associations between QoL domains and measures of Impairment and Perception (See above). METHODS Seventy-Five males, including MD classifications DMD, BMD, LGMD, FSHD and CTRL, completed measures for QoL, Knee-Extension Maximal Voluntary Contraction (KEMVC), Fatigue, Pain, Self-Efficacy and Activities of Daily Living (ADL). RESULTS QoL was lower across many domains in MD than CTRL. FSHD scored lower than DMD for mental wellbeing domains. KEMVC associated with Physical-Function domain for BMD. Pain, Self-Efficacy and ADLs associated with QoL domains, with Fatigue the most consistently associated. CONCLUSION The present study identified differences between MD classifications within self-perceptions of mental-health. Muscle weakness is a defining feature of MD; however, it doesn't define QoL in adults with MD. A greater understanding of mental wellbeing, independence, and management of fatigue and pain, are required to improve QoL for adults with MD.
Collapse
Affiliation(s)
- Matthew F. Jacques
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | | | - Gladys L. Onambele-Pearson
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - Neil D. Reeves
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - Georgina K. Stebbings
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| | - Ellen A. Dawson
- Research Institute for Sport and Exercise Science, Liverpool John Moores University, Liverpool, UK
| | | | - Christopher I. Morse
- Musculoskeletal Science & Sports Medicine Research Centre, School of Healthcare Science, Faculty of Science and Engineering, Manchester Metropolitan University, Manchester, UK
| |
Collapse
|
19
|
Nauman F, Hussain MFA, Burakgazi AZ. The development of myasthenia gravis in a patient with facioscapulohumeral muscular dystrophy: case report and literature review. Neurol Int 2019; 11:8191. [PMID: 31579125 PMCID: PMC6763750 DOI: 10.4081/ni.2019.8191] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 06/10/2019] [Indexed: 11/23/2022] Open
Abstract
The co-existence of facioscapulohumeral muscle dystrophy (FSHD) and myasthenia gravis (MG) is very rare and few cases have been described in the literature. To increase the awareness of the health care providers, we present herein a rare case of MG in a patient with FSHD, discuss the diagnostic challenges, pre- and post-treatment findings and provide a literature review.
Collapse
Affiliation(s)
- Feryal Nauman
- Neuroscience Section, Department of Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| | | | - Ahmet Z Burakgazi
- Neuroscience Section, Department of Medicine, Virginia Tech Carilion School of Medicine, Roanoke, VA, USA
| |
Collapse
|
20
|
Saitoh M, Ishida J, Ebner N, Anker SD, Von Haehling S. Myostatin inhibitors as pharmacological treatment for muscle wasting and muscular dystrophy. JCSM CLINICAL REPORTS 2017. [DOI: 10.17987/jcsm-cr.v2i1.37] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Myostatin, a member of the transforming growth factor beta (TGF-β) superfamily that is highly expressed in skeletal muscle, was first described in 1997. It has been known that loss of myostatin function induces an increase in muscle mass in mice, cow, dogs and humans. Therefore, myostatin and its receptor have emerged as a therapeutic target for loss of skeletal muscle such as sarcopenia and cachexia, as well as muscular dystrophies. At the molecular level, myostatin binds to and activates the activin receptor IIB (ActRIIB)/Alk 4/5 complex. Therapeutic approaches therefore are being taken both pre-clinically and clinically to inhibit the myostatin signaling pathway. Several myostatin inhibitors , including myostatin antibodies, anti-myostatin peptibody, activin A antibody, soluble (decoy) forms of ActRIIB (ActRⅡB-Fc), anti-myostatin adnectin, ActRⅡB antibody have been tested in the last decade. The current review covers the present knowledge of several myostatin inhibitors as therapeutic approach for patients with loss of skeletal muscle however, the available information about compounds in development is limited.
Collapse
|
21
|
Denny AP, Heather AK. Are Antioxidants a Potential Therapy for FSHD? A Review of the Literature. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7020295. [PMID: 28690764 PMCID: PMC5485364 DOI: 10.1155/2017/7020295] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 04/27/2017] [Accepted: 05/03/2017] [Indexed: 11/21/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an inherited myopathy affecting approximately 1 in 7500 individuals worldwide. It is a progressive disease characterised by skeletal muscle weakness and wasting. A genetic mutation on the 4q35 chromosome results in the expression of the double homeobox 4 gene (DUX4) which drives oxidative stress, inflammation, toxicity, and atrophy within the skeletal muscle. FSHD is characterised by oxidative stress, and there is currently no cure and a lack of therapies for the disease. Antioxidants have been researched for many years, with investigators aiming to use antioxidants therapeutically for oxidative stress-associated diseases. This has included both natural and synthetic antioxidants. The use of antioxidants in preclinical or clinical models has been largely successful with a plethora of research reporting positive results. However, when translated to clinical trials, the use of antioxidants as a therapeutic intervention for a variety of disease has been largely unsuccessful. Moreover, specifically focusing on FSHD, limited research has been conducted on the use of antioxidants as a therapy in either preclinical or clinical models. This review summarises the current state of antioxidant use in the treatment of FSHD and discusses their potential avenue for therapeutic use for FSHD patients.
Collapse
Affiliation(s)
- Adam Philip Denny
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| | - Alison Kay Heather
- Department of Physiology, School of Biomedical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
22
|
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
|
23
|
Mnatsakanian A, Kissel JT, Terry P, King WM. One clinic's experience with carbon fiber orthoses in neuromuscular disease. Muscle Nerve 2016; 55:202-205. [PMID: 27356926 DOI: 10.1002/mus.25233] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 06/20/2016] [Accepted: 06/28/2016] [Indexed: 11/06/2022]
Abstract
INTRODUCTION The purpose of this study was to summarize our experience with off-the-shelf anterior shell carbon fiber ankle-foot orthoses (CFAFOs) prescribed to adult neuromuscular patients in an outpatient clinic. METHODS We studied ambulatory patients who were seen in Muscular Dystrophy Association or amyotrophic lateral sclerosis clinics between 2011 and 2014 and prescribed anterior shell CFAFOs. Charts were reviewed with attention to diagnosis, satisfaction with use, and reasons for acceptance or rejection. We included individuals who were currently using AFOs and those being prescribed AFOs for the first time. We were especially interested in reasons for acceptance or rejection of the orthosis. RESULTS Two hundred eighty-three charts were reviewed. Of these, 109 of 123 (89%) patients were satisfied or extremely satisfied with the anterior shell CFAFOs, including 38 who had previously used other styles. CONCLUSION Anterior shell CFAFOs should be considered for most neuromuscular patients with distal leg weakness. Muscle Nerve 55: 202-205, 2017.
Collapse
Affiliation(s)
- Ani Mnatsakanian
- Department of Neurology, The Ohio State University, 395 West 12th Avenue, 7th Floor, Columbus, Ohio, 43210, USA
| | - John T Kissel
- Department of Neurology, The Ohio State University, 395 West 12th Avenue, 7th Floor, Columbus, Ohio, 43210, USA
| | - Philip Terry
- Department of Neurology, The Ohio State University, 395 West 12th Avenue, 7th Floor, Columbus, Ohio, 43210, USA
| | - Wendy M King
- Department of Neurology, The Ohio State University, 395 West 12th Avenue, 7th Floor, Columbus, Ohio, 43210, USA
| |
Collapse
|
24
|
Bergsma A, Cup EHC, Janssen MMHP, Geurts ACH, de Groot IJM. Upper limb function and activity in people with facioscapulohumeral muscular dystrophy: a web-based survey. Disabil Rehabil 2016; 39:236-243. [PMID: 26942834 DOI: 10.3109/09638288.2016.1140834] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Purpose To investigate the upper extremity (UE) at the level of impairments and related activity limitations and participation restrictions in people with facioscapulohumeral muscular dystrophy (FSHD). Methods The study was conducted using web-based questionnaires that were distributed amongst people with FSHD in the Netherlands. Eighty-eight respondents started the survey, and 71 completed it. The questionnaires covered the following dimensions: Function, Activity and Participation of the International Classification of Functioning Disability and Health. Results More than 40% of the respondents experienced pain in one arm or both the arms. Increased pain and stiffness scores and longer disease duration were associated with increased limitation scores. For basic activities, lifting the arm above shoulder-level was most frequently reported as most limited, coherent with the clinical picture of FSHD. Among the respondents, 50% indicated restrictions at school, 78% indicated restrictions at work and more than 80% indicated restrictions whilst participating in sports, hobbies, household activities and romantic relationships. Conclusions This study has shown that alongside the well-known problem of lifting the arms above shoulder-level, UE activities below shoulder height during vocational and occupational activities are also problematic in patients with FSHD. Alongside disease duration, pain and stiffness are associated with UE activity limitations. Implications for Rehabilitation Attention is needed for pain and experienced stiffness in the upper extremity as it is frequently present in patients with FSHD. Rehabilitation professionals need to be aware that patients with FSHD not only experience problems with activities above shoulder height, but also with activities below shoulder height. At least 50% of the patients with FSHD experience restrictions in participation as a result of limitations in their UE.
Collapse
Affiliation(s)
- Arjen Bergsma
- a Department of Rehabilitation , Donders Centre for Neuroscience, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Edith H C Cup
- a Department of Rehabilitation , Donders Centre for Neuroscience, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Mariska M H P Janssen
- a Department of Rehabilitation , Donders Centre for Neuroscience, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Alexander C H Geurts
- a Department of Rehabilitation , Donders Centre for Neuroscience, Radboud University Medical Center , Nijmegen , The Netherlands
| | - Imelda J M de Groot
- a Department of Rehabilitation , Donders Centre for Neuroscience, Radboud University Medical Center , Nijmegen , The Netherlands
| |
Collapse
|
25
|
Dori Z, Sarig Bahat H. Unusual scapular winging - A case report. ACTA ACUST UNITED AC 2016; 24:75-80. [PMID: 26759220 DOI: 10.1016/j.math.2015.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2015] [Revised: 12/06/2015] [Accepted: 12/13/2015] [Indexed: 11/19/2022]
Abstract
Scapular mobility has a central role in maintaining normal upper limb function. Scapular winging is characterized by a failure in the dynamic stabilization of the scapula against the thoracic wall resulting in a condition in which the medial border of the scapula is prominent. The following case describes a patient who was referred to physiotherapy due to abnormal scapular protrusion. The main findings of the physical examination showed weakness of the scapular stabilizers more prominent on the right side than of the left. Additionally, the physical examination demonstrated weakness of the abdominal muscles, hip adductors, and ankle dorsi-flexors, as well as some facial muscles. The electromyography results were inconclusive. Further examination led to clinical suspicion of Facioscapulohumeral Dystrophy (FSHD) as a diagnosis, which was confirmed by genetic testing. Facioscapulohumeral Dystrophy is characterized by symptoms related to motor function and in most cases becomes evident in patients in their 20s and 30s. The disease signs and symptoms are often identified in a clinical setting. Currently, there are no reports describing an effective treatment for the disease. However, physiotherapy, moderate physical exercise, counselling, and use of suitable aids and orthoses may help improve functionality and mobility. This case report aims to increase the awareness of musculoskeletal physiotherapists to this unique dystrophy, when encountering complex presentations with scapular winging.
Collapse
Affiliation(s)
- Zohar Dori
- Department of Physical Therapy, Faculty of Social Welfare & Health Sciences, University of Haifa, Haifa 31905, Israel
| | - Hilla Sarig Bahat
- Department of Physical Therapy, Faculty of Social Welfare & Health Sciences, University of Haifa, Haifa 31905, Israel.
| |
Collapse
|
26
|
Santos DB, Boussaid G, Stojkovic T, Orlikowski D, Letilly N, Behin A, Butel S, Lofaso F, Prigent H. Respiratory muscle dysfunction in facioscapulohumeral muscular dystrophy. Neuromuscul Disord 2015; 25:632-9. [DOI: 10.1016/j.nmd.2015.04.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Revised: 04/20/2015] [Accepted: 04/23/2015] [Indexed: 10/23/2022]
|
27
|
Clinical Muscle Testing Compared with Whole-Body Magnetic Resonance Imaging in Facio-scapulo-humeral Muscular Dystrophy. Clin Neuroradiol 2015; 26:445-455. [DOI: 10.1007/s00062-015-0386-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/13/2015] [Indexed: 12/30/2022]
|
28
|
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
|
29
|
Jones TI, King OD, Himeda CL, Homma S, Chen JCJ, Beermann ML, Yan C, Emerson CP, Miller JB, Wagner KR, Jones PL. Individual epigenetic status of the pathogenic D4Z4 macrosatellite correlates with disease in facioscapulohumeral muscular dystrophy. Clin Epigenetics 2015; 7:37. [PMID: 25904990 PMCID: PMC4405830 DOI: 10.1186/s13148-015-0072-6] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2014] [Accepted: 03/11/2015] [Indexed: 12/27/2022] Open
Abstract
Background Both forms of facioscapulohumeral muscular dystrophy (FSHD) are associated with aberrant epigenetic regulation of the chromosome 4q35 D4Z4 macrosatellite. Chromatin changes due to large deletions of heterochromatin (FSHD1) or mutations in chromatin regulatory proteins (FSHD2) lead to relaxation of epigenetic repression and increased expression of the deleterious double homeobox 4 (DUX4) gene encoded within the distal D4Z4 repeat. However, many individuals with the genetic requirements for FSHD remain asymptomatic throughout their lives. Here we investigated family cohorts of FSHD1 individuals who were either affected (manifesting) or without any discernible weakness (nonmanifesting/asymptomatic) and their unaffected family members to determine if individual epigenetic status and stability of repression at the contracted 4q35 D4Z4 array in myocytes correlates with FSHD disease. Results Family cohorts were analyzed for DNA methylation on the distal pathogenic 4q35 D4Z4 repeat on permissive A-type subtelomeres. We found DNA hypomethylation in FSHD1-affected subjects, hypermethylation in healthy controls, and distinctly intermediate levels of methylation in nonmanifesting subjects. We next tested if these differences in DNA methylation had functional relevance by assaying DUX4-fl expression and the stability of epigenetic repression of DUX4-fl in myogenic cells. Treatment with drugs that alter epigenetic status revealed that healthy cells were refractory to treatment, maintaining stable repression of DUX4, while FSHD1-affected cells were highly responsive to treatment and thus epigenetically poised to express DUX4. Myocytes from nonmanifesting subjects had significantly higher levels of DNA methylation and were more resistant to DUX4 activation in response to epigenetic drug treatment than cells from FSHD1-affected first-degree relatives containing the same contraction, indicating that the epigenetic status of the contracted D4Z4 array is reflective of disease. Conclusions The epigenetic status of the distal 4qA D4Z4 repeat correlates with FSHD disease; FSHD-affected subjects have hypomethylation, healthy unaffected subjects have hypermethylation, and nonmanifesting subjects have characteristically intermediate methylation. Thus, analysis of DNA methylation at the distal D4Z4 repeat could be used as a diagnostic indicator of developing clinical FSHD. In addition, the stability of epigenetic repression upstream of DUX4 expression is a key regulator of disease and a viable therapeutic target. Electronic supplementary material The online version of this article (doi:10.1186/s13148-015-0072-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Takako I Jones
- Department of Neurology and Department of Cell and Developmental Biology, The Wellstone Program, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA
| | - Oliver D King
- Department of Neurology and Department of Cell and Developmental Biology, The Wellstone Program, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA ; The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, 31 Center Drive, Bethesda, MD USA
| | - Charis L Himeda
- Department of Neurology and Department of Cell and Developmental Biology, The Wellstone Program, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA ; The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, 31 Center Drive, Bethesda, MD USA
| | - Sachiko Homma
- Neuromuscular Biology & Disease Group, Departments of Neurology and Physiology & Biophysics, Boston University School of Medicine, 72 E Concord St, Boston, MA 02118 USA
| | - Jennifer C J Chen
- Department of Neurology and Department of Cell and Developmental Biology, The Wellstone Program, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA ; The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, 31 Center Drive, Bethesda, MD USA
| | - Mary Lou Beermann
- Neuromuscular Biology & Disease Group, Departments of Neurology and Physiology & Biophysics, Boston University School of Medicine, 72 E Concord St, Boston, MA 02118 USA
| | - Chi Yan
- Department of Neurology and Department of Cell and Developmental Biology, The Wellstone Program, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA ; Key Lab of Swine Genetics and Breeding, Ministry of Agriculture, College of Animal Science and Technology, Huazhong Agricultural University, No.1, Shizishan Street, Wuhan, 430070 People's Republic of China
| | - Charles P Emerson
- Department of Neurology and Department of Cell and Developmental Biology, The Wellstone Program, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA ; The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, 31 Center Drive, Bethesda, MD USA
| | - Jeffrey B Miller
- Neuromuscular Biology & Disease Group, Departments of Neurology and Physiology & Biophysics, Boston University School of Medicine, 72 E Concord St, Boston, MA 02118 USA
| | - Kathryn R Wagner
- The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, 31 Center Drive, Bethesda, MD USA ; The Hugo W. Moser Research Institute, Kennedy Krieger Institute, and the Departments of Neurology and Neuroscience, The Johns Hopkins School of Medicine, 733 N Broadway, Baltimore, MD 21205 USA
| | - Peter L Jones
- Department of Neurology and Department of Cell and Developmental Biology, The Wellstone Program, University of Massachusetts Medical School, 55 Lake Avenue North, Worcester, MA 01655 USA ; The Eunice Kennedy Shriver National Institute of Child Health and Human Development, Sen. Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, 31 Center Drive, Bethesda, MD USA
| |
Collapse
|
30
|
Lek A, Rahimov F, Jones PL, Kunkel LM. Emerging preclinical animal models for FSHD. Trends Mol Med 2015; 21:295-306. [PMID: 25801126 DOI: 10.1016/j.molmed.2015.02.011] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 02/23/2015] [Accepted: 02/25/2015] [Indexed: 12/18/2022]
Abstract
Facioscapulohumeral dystrophy (FSHD) is a unique and complex genetic disease that is not entirely solved. Recent advances in the field have led to a consensus genetic premise for the disorder, enabling researchers to now pursue the design of preclinical models. In this review we explore all available FSHD models (DUX4-dependent and -independent) for their utility in therapeutic discovery and potential to yield novel disease insights. Owing to the complex nature of FSHD, there is currently no single model that accurately recapitulates the genetic and pathophysiological spectrum of the disorder. Existing models emphasize only specific aspects of the disease, highlighting the need for more collaborative research and novel paradigms to advance the translational research space of FSHD.
Collapse
Affiliation(s)
- Angela Lek
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; The Wellstone Program, Departments of Neurology and Cell and Developmental Biology, University of Massachusetts Medical School (UMMS), Worcester, MA 01655, USA.
| | - Fedik Rahimov
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; The Wellstone Program, Departments of Neurology and Cell and Developmental Biology, University of Massachusetts Medical School (UMMS), Worcester, MA 01655, USA
| | - Peter L Jones
- The Wellstone Program, Departments of Neurology and Cell and Developmental Biology, University of Massachusetts Medical School (UMMS), Worcester, MA 01655, USA; The Eunice Kennedy Shriver National Institute of Child Health and Human Development (NIHCD) Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Louis M Kunkel
- Division of Genetics and Genomics, Manton Center for Orphan Disease Research, Children's Hospital, Boston, MA 02115, USA; Department of Genetics, Harvard Medical School, Boston, MA 02115, USA; The Wellstone Program, Departments of Neurology and Cell and Developmental Biology, University of Massachusetts Medical School (UMMS), Worcester, MA 01655, USA
| |
Collapse
|
31
|
Diagnostic approach for FSHD revisited: SMCHD1 mutations cause FSHD2 and act as modifiers of disease severity in FSHD1. Eur J Hum Genet 2014; 23:808-16. [PMID: 25370034 DOI: 10.1038/ejhg.2014.191] [Citation(s) in RCA: 82] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2014] [Revised: 08/13/2014] [Accepted: 08/19/2014] [Indexed: 11/08/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant muscular disorder with a wide clinical variability. Contractions of the D4Z4 macrosatellite repeat on chromosome 4q35 are the molecular basis of the pathophysiology. Recently, in a subset of patients without D4Z4 repeat contractions, variants in the SMCHD1 gene have been identified that lead to hypomethylation of D4Z4 and thus DUX4 transcription, which causes FSHD type 2. In this study, we have screened 55 FSHD1-negative and 40 FSHD1-positive patients from unrelated families for potentially pathogenic variants in SMCHD1 by next-generation sequencing (NGS). We identified variants in SMCHD1 in 11 index patients, including missense, splice site and non-sense mutations. We developed a pyrosequencing assay to determine the methylation status of the D4Z4 repeat array and found significantly lower methylation levels for FSHD2 patients than for healthy controls and FSHD1 patients. Two out of eleven SMCHD1 mutation carriers had moderately contracted D4Z4 alleles thus these patients are suffering from FSHD1 and 2. Comparing the phenotype of patients, all FSHD2 patients were relatively mildly affected while patients with FSHD1+2 were much more severely affected than expected from their D4Z4 copy number. Our findings confirm the role of SMCHD1 mutations in FSHD2 and as a modifier of disease severity. With SMCHD1 variants found in 16.4% of phenotypic FSHD patients without D4Z4 repeat contractions, the incidence of FSHD2 is rather high and hence we suggest including sequencing of SMCHD1, haplotyping and methylation analysis in the workflow of molecular FSHD diagnostics.
Collapse
|
32
|
Ferri G, Huichalaf CH, Caccia R, Gabellini D. Direct interplay between two candidate genes in FSHD muscular dystrophy. Hum Mol Genet 2014; 24:1256-66. [PMID: 25326393 PMCID: PMC4321439 DOI: 10.1093/hmg/ddu536] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is one of the most common neuromuscular disorders. The major form of the disease (FSHD1) is linked to decrease in copy number of a 3.3-kb tandem repeated macrosatellite (D4Z4), located on chromosome 4q35. D4Z4 deletion alters chromatin structure of the locus leading to aberrant expression of nearby 4q35 genes. Given the high variability in disease onset and progression, multiple factors could contribute to the pathogenesis of FSHD. Among the FSHD candidate genes are double homeobox 4 (DUX4), encoded by the most telomeric D4Z4 unit, and FSHD region gene 1 (FRG1). DUX4 is a sequence-specific transcription factor. Here, we located putative DUX4 binding sites in the human FRG1 genomic area and we show specific DUX4 association to these regions. We found also that ectopically expressed DUX4 up-regulates the endogenous human FRG1 gene in healthy muscle cells, while DUX4 knockdown leads to a decrease in FRG1 expression in FSHD muscle cells. Moreover, DUX4 binds directly and specifically to its binding site located in the human FRG1 gene and transactivates constructs containing FRG1 genomic regions. Intriguingly, the mouse Frg1 genomic area lacks DUX4 binding sites and DUX4 is unable to activate the endogenous mouse Frg1 gene providing a possible explanation for the lack of muscle phenotype in DUX4 transgenic mice. Altogether, our results demonstrate that FRG1 is a direct DUX4 transcriptional target uncovering a novel regulatory circuit contributing to FSHD.
Collapse
Affiliation(s)
- Giulia Ferri
- Division of Regenerative Medicine, Stem Cells, and Gene Therapy, Dulbecco Telethon Institute at San Raffaele Scientific Institute, DIBIT2, 5A3, Via Olgettina 58, 20132 Milan, Italy Università Vita-Salute San Raffaele, Milan, Italy
| | - Claudia H Huichalaf
- Division of Regenerative Medicine, Stem Cells, and Gene Therapy, Dulbecco Telethon Institute at San Raffaele Scientific Institute, DIBIT2, 5A3, Via Olgettina 58, 20132 Milan, Italy Università Vita-Salute San Raffaele, Milan, Italy
| | - Roberta Caccia
- Division of Regenerative Medicine, Stem Cells, and Gene Therapy, Dulbecco Telethon Institute at San Raffaele Scientific Institute, DIBIT2, 5A3, Via Olgettina 58, 20132 Milan, Italy
| | - Davide Gabellini
- Division of Regenerative Medicine, Stem Cells, and Gene Therapy, Dulbecco Telethon Institute at San Raffaele Scientific Institute, DIBIT2, 5A3, Via Olgettina 58, 20132 Milan, Italy
| |
Collapse
|
33
|
Upper Extremity Kinematics and Muscle Activation Patterns in Subjects With Facioscapulohumeral Dystrophy. Arch Phys Med Rehabil 2014; 95:1731-41. [DOI: 10.1016/j.apmr.2014.03.033] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Accepted: 03/27/2014] [Indexed: 10/25/2022]
|
34
|
Bergsma A, Cup EHC, Geurts ACH, de Groot IJM. Upper extremity function and activity in facioscapulohumeral dystrophy and limb-girdle muscular dystrophies: a systematic review. Disabil Rehabil 2014; 37:1017-32. [PMID: 25098592 DOI: 10.3109/09638288.2014.948138] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
PURPOSE The aims of this review were (1) to provide insight into the natural course of upper-extremity (UE) impairments and UE activity limitations associated with facioscapulohumeral dystrophy (FSHD) and limb-girdle muscular dystrophies (LGMD), and (2) to provide an overview of outcome measures used to evaluate UE function and activity in patients with FSHD and LGMD. METHODS Scientific literature databases (PubMed, MEDLINE, EMBASE, CINAHL and Cochrane) were searched for relevant publications. INCLUSION CRITERIA (1) studies that included persons with a diagnosis of FSHD or LGMD; and (2) studies that reported the natural course of the UE functions and/or activity with outcome measures at these levels. RESULTS 247 publications were screened, of which 16 fulfilled the selection criteria. Most studies used manual muscle testing (MMT) to evaluate UE function and the Brooke Scale to evaluate UE mobility activities. The clinical picture of UE impairments and limitations of UE activities in FSHD and LGMD patients was highly variable. In general, FSHD and LGMD patients experience difficulty elevating their upper extremities and the execution of tasks takes considerably longer time. CONCLUSIONS The clinical course of UE impairments and activity limitations associated with FSHD and LGMD is difficult to predict due to its high variability. Although measures like MMT and the Brooke Scale are often used, there is a lack of more specific outcome measures to assess UE function and UE capacity and performance in daily life. Measures such as 3D motion analysis and electromyography (EMG) recordings are recommended to provide additional insight in UE function. Questionnaires like the Abilhand are recommended to assess UE capacity and accelerometry to assess UE performance in daily life. IMPLICATIONS FOR REHABILITATION There is a need for specific outcome measures on the level of UE activity. Both the level of capacity and performance should be assessed. Possible outcome measures include 3D motion analysis to assess UE function, questionnaires like the Abilhand to assess UE capacity and accelerometry to assess performance of UE activities in daily life.
Collapse
Affiliation(s)
- Arjen Bergsma
- Department of Rehabilitation, Donders Centre for Neuroscience, Radboud University Medical Center , Nijmegen , The Netherlands and
| | | | | | | |
Collapse
|
35
|
Yao Z, Snider L, Balog J, Lemmers RJLF, Van Der Maarel SM, Tawil R, Tapscott SJ. DUX4-induced gene expression is the major molecular signature in FSHD skeletal muscle. Hum Mol Genet 2014; 23:5342-52. [PMID: 24861551 DOI: 10.1093/hmg/ddu251] [Citation(s) in RCA: 161] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Facioscapulohumeral dystrophy (FSHD) is caused by decreased epigenetic repression of the D4Z4 macrosatellite array and recent studies have shown that this results in the expression of low levels of the DUX4 mRNA in skeletal muscle. Several other mechanisms have been suggested for FSHD pathophysiology and it remains unknown whether DUX4 expression can account for most of the molecular changes seen in FSHD. Since DUX4 is a transcription factor, we used RNA-seq to measure gene expression in muscle cells transduced with DUX4, and in muscle cells and biopsies from control and FSHD individuals. We show that DUX4 target gene expression is the major molecular signature in FSHD muscle together with a gene expression signature consistent with an immune cell infiltration. In addition, one unaffected individual without a known FSHD-causing mutation showed the expression of DUX4 target genes. This individual has a sibling with FSHD and also without a known FSHD-causing mutation, suggesting the presence of an unidentified modifier locus for DUX4 expression and FSHD. These findings demonstrate that the expression of DUX4 accounts for the majority of the gene expression changes in FSHD skeletal muscle together with an immune cell infiltration.
Collapse
Affiliation(s)
- Zizhen Yao
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA 98109, USA
| | - Lauren Snider
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA 98109, USA
| | - Judit Balog
- Department of Human Genetics, Leiden University Medical Center, Leiden, ZA 2333, The Netherlands
| | - Richard J L F Lemmers
- Department of Human Genetics, Leiden University Medical Center, Leiden, ZA 2333, The Netherlands
| | - Silvère M Van Der Maarel
- Department of Human Genetics, Leiden University Medical Center, Leiden, ZA 2333, The Netherlands
| | - Rabi Tawil
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Stephen J Tapscott
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle WA 98109, USA Department of Neurology, University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
36
|
Neguembor MV, Jothi M, Gabellini D. Long noncoding RNAs, emerging players in muscle differentiation and disease. Skelet Muscle 2014; 4:8. [PMID: 24685002 PMCID: PMC3973619 DOI: 10.1186/2044-5040-4-8] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 03/11/2014] [Indexed: 12/18/2022] Open
Abstract
The vast majority of the mammalian genome is transcribed giving rise to many different types of noncoding RNAs. Among them, long noncoding RNAs are the most numerous and functionally versatile class. Indeed, the lncRNA repertoire might be as rich as the proteome. LncRNAs have emerged as key regulators of gene expression at multiple levels. They play important roles in the regulation of development, differentiation and maintenance of cell identity and they also contribute to disease. In this review, we present recent advances in the biology of lncRNAs in muscle development and differentiation. We will also discuss the contribution of lncRNAs to muscle disease with a particular focus on Duchenne and facioscapulohumeral muscular dystrophies.
Collapse
Affiliation(s)
| | | | - Davide Gabellini
- Dulbecco Telethon Institute at San Raffaele Scientific Institute, Division of Regenerative Medicine, Stem cells, and Gene therapy, DIBIT2, 5A3, Via Olgettina 58, 20132 Milano, Italy.
| |
Collapse
|
37
|
Ball AR, Chen YY, Yokomori K. Mechanisms of cohesin-mediated gene regulation and lessons learned from cohesinopathies. BIOCHIMICA ET BIOPHYSICA ACTA 2014; 1839:191-202. [PMID: 24269489 PMCID: PMC3951616 DOI: 10.1016/j.bbagrm.2013.11.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/18/2013] [Revised: 11/09/2013] [Accepted: 11/14/2013] [Indexed: 12/16/2022]
Abstract
Cohesins are conserved and essential Structural Maintenance of Chromosomes (SMC) protein-containing complexes that physically interact with chromatin and modulate higher-order chromatin organization. Cohesins mediate sister chromatid cohesion and cellular long-distance chromatin interactions affecting genome maintenance and gene expression. Discoveries of mutations in cohesin's subunits and its regulator proteins in human developmental disorders, so-called "cohesinopathies," reveal crucial roles for cohesins in development and cellular growth and differentiation. In this review, we discuss the latest findings concerning cohesin's functions in higher-order chromatin architecture organization and gene regulation and new insight gained from studies of cohesinopathies. This article is part of a Special Issue entitled: Chromatin and epigenetic regulation of animal development.
Collapse
Affiliation(s)
- Alexander R Ball
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697-1700, USA
| | - Yen-Yun Chen
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697-1700, USA
| | - Kyoko Yokomori
- Department of Biological Chemistry, School of Medicine, University of California, Irvine, CA 92697-1700, USA.
| |
Collapse
|
38
|
Minis MAH, Satink T, Kinébanian A, Engels JA, Heerkens YF, van Engelen BGM, Nijhuis-van der Sanden MWG. How persons with a neuromuscular disease perceive employment participation: a qualitative study. JOURNAL OF OCCUPATIONAL REHABILITATION 2014; 24:52-67. [PMID: 23645359 DOI: 10.1007/s10926-013-9447-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
INTRODUCTION A qualitative study was carried out to understand how people with a slow progressive adult type neuromuscular disease (NMD) perceive employment participation. METHODS 16 paid employed persons with NMD were interviewed in open, in-depth interviews. Data were analyzed using the constant comparison method. RESULTS Four themes were identified in the analyses: (1) Experiences regarding the meaning of work; (2) Solving problems oneself; (3) Reaching a turning point; and (4) Taking into account environmental aspects. Persons with NMD highlighted benefits of staying at work as well as the tension they felt how to shape decisions to handle progressive physical hindrances in job retention. This study shows how participants at work with NMD were challenged to keep up appearances at work and at home, the tension felt around when and if to disclose, the effect of their condition on colleagues and work reorganisation challenges. Participants experienced that disclosure did not always make things better. With increasing disability participants' focus shifted from the importance of assistive products towards considerate colleague, in particular superior's willingness in supporting job retention. CONCLUSIONS Implications for health professionals might include awareness of the significant impact of changes in physical condition on employment. Timely communication and if appropriate referral to a health or occupational professional may empower employees with NMD to handle employment issues at a for themselves appropriate way. Assistive products and a supportive superior might enhance employment participation.
Collapse
Affiliation(s)
- Marie-Antoinette H Minis
- Department of Occupation and Health, HAN University of Applied Sciences, PO Box 6960, 6503 GL, Nijmegen, The Netherlands,
| | | | | | | | | | | | | |
Collapse
|
39
|
Janssen BH, Voet NBM, Nabuurs CI, Kan HE, de Rooy JWJ, Geurts AC, Padberg GW, van Engelen BGM, Heerschap A. Distinct disease phases in muscles of facioscapulohumeral dystrophy patients identified by MR detected fat infiltration. PLoS One 2014; 9:e85416. [PMID: 24454861 PMCID: PMC3891814 DOI: 10.1371/journal.pone.0085416] [Citation(s) in RCA: 120] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2013] [Accepted: 11/26/2013] [Indexed: 11/18/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an untreatable disease, characterized by asymmetric progressive weakness of skeletal muscle with fatty infiltration. Although the main genetic defect has been uncovered, the downstream mechanisms causing FSHD are not understood. The objective of this study was to determine natural disease state and progression in muscles of FSHD patients and to establish diagnostic biomarkers by quantitative MRI of fat infiltration and phosphorylated metabolites. MRI was performed at 3T with dedicated coils on legs of 41 patients (28 men/13 women, age 34-76 years), of which eleven were re-examined after four months of usual care. Muscular fat fraction was determined with multi spin-echo and T1 weighted MRI, edema by TIRM and phosphorylated metabolites by 3D (31)P MR spectroscopic imaging. Fat fractions were compared to clinical severity, muscle force, age, edema and phosphocreatine (PCr)/ATP. Longitudinal intramuscular fat fraction variation was analyzed by linear regression. Increased intramuscular fat correlated with age (p<0.05), FSHD severity score (p<0.0001), inversely with muscle strength (p<0.0001), and also occurred sub-clinically. Muscles were nearly dichotomously divided in those with high and with low fat fraction, with only 13% having an intermediate fat fraction. The intramuscular fat fraction along the muscle's length, increased from proximal to distal. This fat gradient was the steepest for intermediate fat infiltrated muscles (0.07±0.01/cm, p<0.001). Leg muscles in this intermediate phase showed a decreased PCr/ATP (p<0.05) and the fastest increase in fatty infiltration over time (0.18±0.15/year, p<0.001), which correlated with initial edema (p<0.01), if present. Thus, in the MR assessment of fat infiltration as biomarker for diseased muscles, the intramuscular fat distribution needs to be taken into account. Our results indicate that healthy individual leg muscles become diseased by entering a progressive phase with distal fat infiltration and altered energy metabolite levels. Fat replacement then relatively rapidly spreads over the whole muscle.
Collapse
Affiliation(s)
- Barbara H. Janssen
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
- * E-mail:
| | - Nicoline B. M. Voet
- Department of Rehabilitation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Christine I. Nabuurs
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hermien E. Kan
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Jacky W. J. de Rooy
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alexander C. Geurts
- Department of Rehabilitation, Radboud University Medical Center, Nijmegen, The Netherlands
| | - George W. Padberg
- Department of Neurology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Arend Heerschap
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
40
|
Lassche S, Ottenheijm CAC, Voermans NC, Westeneng HJ, Janssen BH, van der Maarel SM, Hopman MT, Padberg GW, Stienen GJM, van Engelen BGM. Determining the role of sarcomeric proteins in facioscapulohumeral muscular dystrophy: a study protocol. BMC Neurol 2013; 13:144. [PMID: 24119284 PMCID: PMC3852245 DOI: 10.1186/1471-2377-13-144] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2013] [Accepted: 10/02/2013] [Indexed: 12/26/2022] Open
Abstract
Background Although muscle weakness is a hallmark of facioscapulohumeral muscular dystrophy (FSHD), the molecular mechanisms that lead to weakness in FSHD remain largely unknown. Recent studies suggest aberrant expression of genes involved in skeletal muscle development and sarcomere contractility, and activation of pathways involved in sarcomeric protein degradation. This study will investigate the contribution of sarcomeric protein dysfunction to the pathogenesis of muscle weakness in FSHD. Methods/Design Evaluation of sarcomeric function using skinned single muscle fiber contractile studies and protein analysis in muscle biopsies (quadriceps femoris and tibialis anterior) from patients with FSHD and age- and gender-matched healthy controls. Patients with other forms of muscular dystrophy and inflammatory myopathy will be included as disease controls to assess whether results are due to changes specific for FSHD, or a consequence of muscle disease in general. A total of 56 participants will be included. Extensive clinical parameters will be measured using MRI, quantitative muscle studies and physical activity assessments. Discussion This study is the first to extensively investigate muscle fiber physiology in FSHD following an earlier pilot study suggesting sarcomeric dysfunction in FSHD. The results obtained in this study will increase the understanding of the pathophysiology of muscle weakness in FSHD, and possibly identify novel targets for therapeutic intervention.
Collapse
Affiliation(s)
- Saskia Lassche
- Department of Neurology, Radboud university medical center, Nijmegen, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Sacconi S, Lemmers R, Balog J, van der Vliet P, Lahaut P, van Nieuwenhuizen M, Straasheijm K, Debipersad R, Vos-Versteeg M, Salviati L, Casarin A, Pegoraro E, Tawil R, Bakker E, Tapscott S, Desnuelle C, van der Maarel S. The FSHD2 gene SMCHD1 is a modifier of disease severity in families affected by FSHD1. Am J Hum Genet 2013; 93:744-51. [PMID: 24075187 DOI: 10.1016/j.ajhg.2013.08.004] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2013] [Revised: 07/04/2013] [Accepted: 08/01/2013] [Indexed: 01/04/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy type 1 (FSHD1) is caused by contraction of the D4Z4 repeat array on chromosome 4 to a size of 1-10 units. The residual number of D4Z4 units inversely correlates with clinical severity, but significant clinical variability exists. Each unit contains a copy of the DUX4 retrogene. Repeat contractions are associated with changes in D4Z4 chromatin structure that increase the likelihood of DUX4 expression in skeletal muscle, but only when the repeat resides in a genetic background that contains a DUX4 polyadenylation signal. Mutations in the structural maintenance of chromosomes flexible hinge domain containing 1 (SMCHD1) gene, encoding a chromatin modifier of D4Z4, also result in the increased likelihood of DUX4 expression in individuals with a rare form of FSHD (FSHD2). Because SMCHD1 directly binds to D4Z4 and suppresses somatic expression of DUX4, we hypothesized that SMCHD1 may act as a genetic modifier in FSHD1. We describe three unrelated individuals with FSHD1 presenting an unusual high clinical severity based on their upper-sized FSHD1 repeat array of nine units. Each of these individuals also carries a mutation in the SMCHD1 gene. Familial carriers of the FSHD1 allele without the SMCHD1 mutation were only mildly affected, suggesting a modifier effect of the SMCHD1 mutation. Knocking down SMCHD1 in FSHD1 myotubes increased DUX4 expression, lending molecular support to a modifier role for SMCHD1 in FSHD1. We conclude that FSHD1 and FSHD2 share a common pathophysiological pathway in which the FSHD2 gene can act as modifier for disease severity in families affected by FSHD1.
Collapse
|
42
|
Stadler G, Rahimov F, King OD, Chen JCJ, Robin JD, Wagner KR, Shay JW, Emerson CP, Wright WE. Telomere position effect regulates DUX4 in human facioscapulohumeral muscular dystrophy. Nat Struct Mol Biol 2013; 20:671-8. [PMID: 23644600 PMCID: PMC3711615 DOI: 10.1038/nsmb.2571] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2012] [Accepted: 03/19/2013] [Indexed: 11/09/2022]
Abstract
Telomeres may regulate human disease by at least two independent mechanisms. First, replicative senescence occurs once short telomeres generate DNA-damage signals that produce a barrier to tumor progression. Second, telomere position effects (TPE) could change gene expression at intermediate telomere lengths in cultured human cells. Here we report that telomere length may contribute to the pathogenesis of facioscapulohumeral muscular dystrophy (FSHD). FSHD is a late-onset disease genetically residing only 25-60 kilobases from the end of chromosome 4q. We used a floxable telomerase to generate isogenic clones with different telomere lengths from affected patients and their unaffected siblings. DUX4, the primary candidate for FSHD pathogenesis, is upregulated over ten-fold in FSHD myoblasts and myotubes with short telomeres, and its expression is inversely proportional to telomere length. FSHD may be the first known human disease in which TPE contributes to age-related phenotype.
Collapse
Affiliation(s)
- Guido Stadler
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas,
TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| | - Fedik Rahimov
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
- Program in Genomics, Division of Genetics, Boston Children’s Hospital,
Harvard Medical School, Boston, MA 02115
| | - Oliver D. King
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
- Boston Biomedical Research Institute, Watertown, MA 02472
| | - Jennifer C. J. Chen
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
- Boston Biomedical Research Institute, Watertown, MA 02472
| | - Jerome D. Robin
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas,
TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| | - Kathryn R. Wagner
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
- The Hugo W. Moser Research Institute at Kennedy Krieger Institute, Baltimore, MD
21205
- Department of Neurology and Neuroscience, Johns Hopkins School of Medicine,
Baltimore, MD 21205
| | - Jerry W. Shay
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas,
TX 75390
- CEGMR, King Abulaziz University, Jeddah, Saudi Arabia
| | - Charles P. Emerson
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
- Boston Biomedical Research Institute, Watertown, MA 02472
| | - Woodring E. Wright
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas,
TX 75390
- Senator Paul D. Wellstone Muscular Dystrophy Cooperative Research Center
| |
Collapse
|
43
|
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
|
44
|
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
|
45
|
Giussani M, Cardone MF, Bodega B, Ginelli E, Meneveri R. Evolutionary history of linked D4Z4 and Beta satellite clusters at the FSHD locus (4q35). Genomics 2012; 100:289-96. [PMID: 22824653 PMCID: PMC3488192 DOI: 10.1016/j.ygeno.2012.07.011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Revised: 06/21/2012] [Accepted: 07/12/2012] [Indexed: 01/10/2023]
Abstract
We performed a detailed genomic investigation of the chimpanzee locus syntenic to human chromosome 4q35.2, associated to the facioscapulohumeral dystrophy. Two contigs of approximately 150 kb and 200 kb were derived from PTR chromosomes 4q35 and 3p12, respectively: both regions showed a very similar sequence organization, including D4Z4 and Beta satellite linked clusters. Starting from these findings, we derived a hypothetical evolutionary history of human 4q35, 10q26 and 3p12 chromosome regions focusing on the D4Z4–Beta satellite linked organization. The D4Z4 unit showed an open reading frame (DUX4) at both PTR 4q35 and 3p12 regions; furthermore some subregions of the Beta satellite unit showed a high degree of conservation between chimpanzee and humans. In conclusion, this paper provides evidence that at the 4q subtelomere the linkage between D4Z4 and Beta satellite arrays is a feature that appeared late during evolution and is conserved between chimpanzee and humans.
Collapse
Affiliation(s)
- Marta Giussani
- Department of Biology and Genetics for Medical Sciences, University of Milan, Milan, Italy.
| | | | | | | | | |
Collapse
|
46
|
Neguembor MV, Gabellini D. In junk we trust: repetitive DNA, epigenetics and facioscapulohumeral muscular dystrophy. Epigenomics 2012; 2:271-87. [PMID: 22121874 DOI: 10.2217/epi.10.8] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant myopathy with a peculiar etiology. Unlike most genetic disorders, FSHD is not caused by mutations in a protein-coding gene. Instead, it is associated with contraction of the D4Z4 macrosatellite repeat array located at 4q35. Interestingly, D4Z4 deletion is not sufficient per se to cause FSHD. Moreover, the disease severity, its rate of progression and the distribution of muscle weakness display great variability even among close family relatives. Hence, additional genetic and epigenetic events appear to be required for FSHD pathogenesis. Indeed, recent findings suggest that virtually all levels of epigenetic regulation, from DNA methylation to higher order chromosomal architecture, exhibit alterations in the disease locus causing deregulation of 4q35 gene expression, ultimately leading to FSHD.
Collapse
Affiliation(s)
- Maria V Neguembor
- International PhD Program in Cellular & Molecular Biology, Vita-Salute San Raffaele University, Milan, Italy
| | | |
Collapse
|
47
|
Röthlisberger M, Pioletti DP, Farron A, Terrier A. Effect of a pathological scapular tilt after total shoulder arthroplasty. Comput Methods Biomech Biomed Engin 2012; 16:1196-201. [PMID: 22375919 DOI: 10.1080/10255842.2012.656612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
Total shoulder arthroplasty (TSA) is an accepted and most successfully used treatment for different shoulder pathologies. Different risk factors for the failure of the prosthesis are known. A pathological scapular orientation, observed in elderly people or in patients suffering from neuromuscular diseases, could be a cause of failure, which has not been investigated yet. To test this hypothesis, a numerical musculoskeletal model of the glenohumeral joint was used to compare two TSA cases: a reference normal case and a case with a pathological anterior tilt of the scapula. An active abduction of 150° was simulated. Joint force, contact pattern, polyethylene and cement stress were evaluated for both cases. The pathological tilt slightly increased the joint force and the contact pressure, but also shifted the contact pattern. This eccentric contact increased the stress level within the polyethylene of the glenoid component and within the surrounding cement layer. This adverse effect occurred mainly during the first 60° of abduction. Therefore, a pathological orientation of the scapula may increase the risk of a failure of the cement layer around the glenoid component. These preliminary numerical results should be confirmed by a clinical study.
Collapse
|
48
|
Ramos VFML, Thaisetthawatkul P. A case of fascioscapulohumeral muscular dystrophy misdiagnosed as Becker's muscular dystrophy for 20 years. Age Ageing 2012; 41:273-4. [PMID: 21795275 DOI: 10.1093/ageing/afr095] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
A 60-year-old man diagnosed clinically with Becker's muscular dystrophy 20 years ago by another physician presented with gradually progressive proximal muscle weakness since teenage years. Family history revealed a strong paternal familial inheritance pattern of similar distribution of weakness-face, forearm flexion, knee extension and foot dorsiflexion. Work-ups revealed B12 deficiency and allele 1 deletion in fascioscapulohumeral muscular dystrophy (FSHD) DNA testing. FSHD is the third most common muscular dystrophy. Clinical diagnosis is made from the distinctive pattern of weakness, autosomal-dominant inheritance, and confirmed by genetic testing. This case strongly demonstrates the importance of a thorough and careful clinical evaluation even in a case with a long standing diagnosis.
Collapse
Affiliation(s)
- Vesper Fe Marie Llaneza Ramos
- Neurological Sciences, University of Nebraska Medical Center, 982045 Nebraska Medical Center, Omaha, NE 68198-2045, USA.
| | | |
Collapse
|
49
|
Abstract
Double homeobox 4 (DUX4) is a candidate disease gene for facioscapulohumeral dystrophy (FSHD), one of the most common muscular dystrophies characterized by progressive skeletal muscle degeneration. Despite great strides in understanding precise genetics of FSHD, the molecular pathophysiology of the disease remains unclear. One of the major limitations has been the availability of appropriate molecular tools to study DUX4 protein. In the present study, we report the development of five new monoclonal antibodies targeted against the N- and C-termini of human DUX4, and characterize their reactivity using Western blot and immunofluorescence staining. Additionally, we show that expression of the canonical full coding DUX4 induces cell death in human primary muscle cells, whereas the expression of a shorter splice form of DUX4 results in no such toxicity. Immunostaining with these new antibodies reveals a differential effect of two DUX4 isoforms on human muscle cells. These antibodies will provide an excellent tool for investigating the role of DUX4 in FSHD pathogenesis.
Collapse
Affiliation(s)
- Linda N Geng
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.
| | | | | |
Collapse
|
50
|
Oygard K, Haestad H, Jørgensen L. Physiotherapy, based on the Bobath concept, may influence the gait pattern in persons with limb-girdle muscle dystrophy: a multiple case series study. PHYSIOTHERAPY RESEARCH INTERNATIONAL 2011; 16:20-31. [PMID: 21110410 DOI: 10.1002/pri.469] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND AND PURPOSE There are few studies on possible effects of physiotherapy for adults with muscular dystrophy. The aim of this study was to examine if treatment based on the Bobath concept may influence specific gait parameters in some of these patients. METHODS A single-subject experimental design with A-B-A-A phases was used, and four patients, three with limb-girdle muscular dystrophy (LGMD) and one with fascioscapulohumeral muscular dystrophy (FSHD), were included. The patients had 1 hour of individually tailored physiotherapy at each working day for a period of 3 weeks. Step length, step width and gait velocity were measured during the A-B-A-A phases by use of an electronic walkway. Walking distance and endurance were measured by use of the '6 minute walk test'. RESULTS . The three LGMD patients, who initially walked with a wide base of support, had a narrower, velocity-adjusted step width after treatment, accompanied with the same or even longer step length. These changes lasted throughout follow-up. Moreover, two of the patients were able to walk a longer distance within 6 minutes after the treatment period. The fourth patient (with FSHD) had a normal step width at baseline, which did not change during the study. CONCLUSIONS The results indicate that physiotherapy treatment based on the Bobath concept may influence the gait pattern in patients with LGMD. However, in order to evaluate the effectiveness of physiotherapy to patients with muscular dystrophies, we call for larger studies and controlled trials.
Collapse
Affiliation(s)
- Kjellaug Oygard
- Section of Physiotherapy, Department of Clinical Therapeutic Services, University Hospital of Northern Norway, Tromsø, Norway.
| | | | | |
Collapse
|