1
|
Kokitsu-Nakata NM, Segarra VCD, Tonello C, Brandão MM, Alonso N, Zechi-Ceide RM. Hemiarhinia caused by a missense variation in SMCHD1: A mild phenotype in the clinical spectrum of Bosma arhinia microphthalmia syndrome. Am J Med Genet A 2024; 194:e63640. [PMID: 38808953 DOI: 10.1002/ajmg.a.63640] [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: 11/04/2022] [Revised: 04/02/2024] [Accepted: 04/11/2024] [Indexed: 05/30/2024]
Abstract
Bosma arhinia microphthalmia syndrome (BAMS, OMIM #603457) is a rare autosomal dominant disorder caused by heterozygous variation in the SMCHD1 gene on chromosome 18p11. Clinically, it is characterized by microphthalmia, absence or hypoplasia of nose, choanal atresia, anosmia, palatal abnormalities, hypogonadotropic hypogonadism, and cryptorchidism. Here we report a Brazilian patient with a likely pathogenic variation in SMCHD1 gene (c.1418A>T; p.Glu473Val) presenting hemiarhinia associated with short stature and hypogonadotropic hypogonadism. Due to the clinical variability of BAMS, we considered that hemiarhinia, without microphthalmia, in the present case, can be considered a mild form of BAMS and could be considered for screening of SMCHD1 gene variation.
Collapse
Affiliation(s)
- Nancy Mizue Kokitsu-Nakata
- Department of Clinical Genetics, Hospital for Rehabilitation of Craniofacial Anomalies (HRAC), University of São Paulo (USP), Bauru, Brazil
- Craniofacial Team, Hospital for Rehabilitation of Craniofacial Anomalies (HRAC), University of São Paulo (USP), Bauru, Brazil
| | - Vinicius Contrucci Dantas Segarra
- Department of Clinical Genetics, Hospital for Rehabilitation of Craniofacial Anomalies (HRAC), University of São Paulo (USP), Bauru, Brazil
| | - Cristiano Tonello
- Craniofacial Team, Hospital for Rehabilitation of Craniofacial Anomalies (HRAC), University of São Paulo (USP), Bauru, Brazil
| | - Michele Madeira Brandão
- Craniofacial Team, Hospital for Rehabilitation of Craniofacial Anomalies (HRAC), University of São Paulo (USP), Bauru, Brazil
| | - Nivaldo Alonso
- Craniofacial Team, Hospital for Rehabilitation of Craniofacial Anomalies (HRAC), University of São Paulo (USP), Bauru, Brazil
| | - Roseli Maria Zechi-Ceide
- Department of Clinical Genetics, Hospital for Rehabilitation of Craniofacial Anomalies (HRAC), University of São Paulo (USP), Bauru, Brazil
| |
Collapse
|
2
|
Sanchez-Sotelo J. Evaluation of the Dysfunctional Scapula. Tech Hand Up Extrem Surg 2024; 28:182-191. [PMID: 39155676 DOI: 10.1097/bth.0000000000000484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Periscapular pain and dysfunction are relatively common complaints in the practice of upper extremity surgeons. However, evaluation of the dysfunctional scapula is intimidating for most. Physical examination of the periscapular muscles is very rich, and a systematic approach provides the opportunity to establish a diagnosis for most patients. Conditions underlying scapular dysfunction include trapezius palsy, serratus palsy, brachial plexus injuries, muscular dystrophy, snapping scapula, pectoralis minor syndrome, congenital undescended scapula, and functional scapular dyskinesis, among others. Patients should be examined with their torso uncovered and assessed from the front and back. Inspection, evaluation of motion, and strength testing may be followed by certain examination maneuvers specific for each condition. The scapular assistance test may help predict the outcome of treatment for certain conditions. Plain radiographs oftentimes need to be complemented with magnetic resonance of the chest to capture periscapular muscles as well as an electromyogram with nerve conduction studies. Computed tomography is particularly helpful for skeletal abnormalities. Ultrasound is used to assess certain peripheral nerve injuries and to guide diagnostic and therapeutic injections. A focused evaluation of the scapula followed by a more specific and directed approach for each condition suspected will allow upper extremity surgeons to approach the evaluation of the dysfunctional scapula with much more confidence.
Collapse
|
3
|
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
|
4
|
Kools J, Voermans N, Jiang JG, Mitelman O, Mellion ML, Ramana V, van Engelen BGM. An open-label pilot study of losmapimod to evaluate the safety, tolerability, and changes in biomarker and clinical outcome assessments in participants with facioscapulohumeral muscular dystrophy type 1. J Neurol Sci 2024; 462:123096. [PMID: 38959779 DOI: 10.1016/j.jns.2024.123096] [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: 01/17/2024] [Revised: 06/05/2024] [Accepted: 06/12/2024] [Indexed: 07/05/2024]
Abstract
INTRODUCTION Facioscapulohumeral muscular dystrophy (FSHD) is a genetic disease caused by aberrant DUX4 expression, leading to progressive muscle weakness. No effective pharmaceutical treatment is available. Losmapimod, a small molecule selective inhibitor of p38 α/β MAPK, showed promising results in a phase 1 trial for the treatment of FSHD, prompting additional studies. We report the findings of an open-label phase 2 trial (NCT04004000) investigating the safety, tolerability, pharmacokinetics, pharmacodynamics, and exploratory efficacy of losmapimod in participants with FSHD1. METHODS This study was conducted at a single site in the Netherlands from August 2019 to March 2021, with an optional, ongoing open-label extension. Participants aged 18 to 65 years with FSHD1 took 15 mg of losmapimod twice daily for 52 weeks. Primary endpoints were measures of losmapimod safety and tolerability. Secondary endpoints were assessments of losmapimod pharmacokinetics and pharmacodynamics. RESULTS Fourteen participants were enrolled. No deaths, serious treatment-emergent adverse events (TEAEs), or discontinuations due to TEAEs were reported. Losmapimod achieved blood concentrations and target engagements that were previously associated with decreased DUX4 expression in vitro. Clinical outcome measures showed a trend toward stabilization or improvement. CONCLUSIONS Losmapimod was well tolerated and may be a promising new treatment for FSHD; a larger phase 3 study is ongoing.
Collapse
Affiliation(s)
- Joost Kools
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Nicol Voermans
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands.
| | | | | | | | | | - Baziel G M van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, the Netherlands.
| |
Collapse
|
5
|
Loureiro M, Branco C, Duarte J, Coutinho G, Martins MM, Novo A. Cardiac Rehabilitation in a Transplanted Person with Emery-Dreifuss Muscular Dystrophy. Arq Bras Cardiol 2023; 120:e20220560. [PMID: 37466619 PMCID: PMC10364993 DOI: 10.36660/abc.20220560] [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: 08/16/2022] [Revised: 03/15/2023] [Accepted: 04/05/2023] [Indexed: 07/20/2023] Open
Abstract
Emery-Dreifuss muscular dystrophy is a rare hereditary neuromuscular disease. Its manifestations begin primarily in childhood. The most frequent manifestations are progressive muscle weakness, atrophy that usually begins in the scapula-vertebral region, extending later to the pelvic girdle, and spinal stiffness. Patients can also manifest cardiac involvement as palpitations, syncope, exercise intolerance, congestive heart failure, and variable heart rhythm disturbances. 1 - 3 The presence and severity of these manifestations can vary according to the individual and the disease's subtypes. 2 Cardiac involvement is the most worrisome feature of this disease, and there are some reports of the need for heart transplantation in this dystrophy. 4.
Collapse
Affiliation(s)
- Maria Loureiro
- Instituto Ciências Biomédicas Abel SalazarPortoPortugalInstituto Ciências Biomédicas Abel Salazar, Porto – Portugal
- Centro Hospitalar e Universitário de CoimbraCoimbraPortugalCentro Hospitalar e Universitário de Coimbra, Coimbra – Portugal
- Cintesis@RISE-NursIDPortoPortugalCintesis@RISE-NursID, Porto – Portugal
| | - Carlos Branco
- Centro Hospitalar e Universitário de CoimbraCoimbraPortugalCentro Hospitalar e Universitário de Coimbra, Coimbra – Portugal
| | - João Duarte
- Centro Hospitalar e Universitário de CoimbraCoimbraPortugalCentro Hospitalar e Universitário de Coimbra, Coimbra – Portugal
| | - Gonçalo Coutinho
- Centro Hospitalar e Universitário de CoimbraCoimbraPortugalCentro Hospitalar e Universitário de Coimbra, Coimbra – Portugal
- Faculdade de Medicina de CoimbraCoimbraPortugalFaculdade de Medicina de Coimbra, Coimbra – Portugal
| | - Maria Manuela Martins
- Instituto Ciências Biomédicas Abel SalazarPortoPortugalInstituto Ciências Biomédicas Abel Salazar, Porto – Portugal
- Cintesis@RISE-NursIDPortoPortugalCintesis@RISE-NursID, Porto – Portugal
- Escola Superior de Enfermagem do PortoPortoPortugalEscola Superior de Enfermagem do Porto, Porto – Portugal
| | - André Novo
- Cintesis@RISE-NursIDPortoPortugalCintesis@RISE-NursID, Porto – Portugal
- Instituto Politécnico de BragançaBragançaPortugalInstituto Politécnico de Bragança, Bragança – Portugal
| |
Collapse
|
6
|
Mondesert E, Bargnoux AS, Portet F, Laoudj-Chenivesse D, Arbogast S, Badiou S, Brun JF, Kuster N, Raynaud de Mauverger E, Cristol JP. Cystatin C for kidney function assessment in patients with facioscapulohumeral muscular dystrophy. Clin Chim Acta 2023; 544:117328. [PMID: 37031784 DOI: 10.1016/j.cca.2023.117328] [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: 02/17/2023] [Revised: 03/28/2023] [Accepted: 03/29/2023] [Indexed: 04/11/2023]
Abstract
BACKGROUND AND AIMS Muscle mass (MM) impairment observed in facioscapulohumeral muscular dystrophy (FSHD) may bias estimated glomerular filtration rate (eGFR) based on creatinine (eGFRcreat). eGFR based on cystatin C (eGFRcys), produced by all nucleated cells, should be an interesting alternative. Main objectives were to compare eGFRcreat and eGRFcys for chronic kidney disease (CKD) staging and for annual eGFR evolution. Secondary objective was to analyse creatinine, cystatin C with measured MM. MATERIAL AND METHODS During 4 years, 159 FSHD patients having one or more creatinine and cystatin C measurements (total samples: n=379), with MM determination by bio-impedancemetry during their follow-up were included. eGFR were determined with CKD-Epi and EKFC equations. RESULTS On first examination samples, mean eGFRcys was significantly lower than mean eGFRcreat of 25.5 and 17.9 ml/min/1.73m2 using CKD-Epi and EKFC equations, respectively. 53.5% (CKD-Epi) and 59.1% (EKFC) of agreement were obtained when using eGFRcys instead of eGFRcreat with reclassifications occurring mainly towards more severe stages. Age was correlated with cystatin C but not with creatinine, MM was correlated with creatinine but not with cystatin C. eGFR decreases >1 ml/min/1.73m2 were more important when using eGFRcys instead of eGFRcreat (CKD-Epi: 37.5 vs 15.4%, p<0.001; EKFC: 34.6 vs 20.2%, p<0.01). CONCLUSION Cystatin C which is independent of MM appears as a promising candidate biomarker for CKD diagnosis and follow-up in FSHD patient.
Collapse
Affiliation(s)
- Etienne Mondesert
- Department of Biochemistry, University Hospital of Montpellier, Montpellier, France
| | - Anne-Sophie Bargnoux
- Department of Biochemistry, University Hospital of Montpellier, Montpellier, France; PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Florence Portet
- Department of Clinical Physiology, University Hospital of Montpellier, France
| | | | - Sandrine Arbogast
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Stéphanie Badiou
- Department of Biochemistry, University Hospital of Montpellier, Montpellier, France; PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Jean-Frédéric Brun
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; Department of Clinical Physiology, University Hospital of Montpellier, France
| | - Nils Kuster
- Department of Biochemistry, University Hospital of Montpellier, Montpellier, France; PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Eric Raynaud de Mauverger
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France; Department of Clinical Physiology, University Hospital of Montpellier, France
| | - Jean-Paul Cristol
- Department of Biochemistry, University Hospital of Montpellier, Montpellier, France; PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France.
| |
Collapse
|
7
|
Emmert D, Szczypien N, Bender TTA, Grigull L, Gass A, Link C, Klawonn F, Conrad R, Mücke M, Sellin J. A diagnostic support system based on pain drawings: binary and k-disease classification of EDS, GBS, FSHD, PROMM, and a control group with Pain2D. Orphanet J Rare Dis 2023; 18:70. [PMID: 36978184 PMCID: PMC10053427 DOI: 10.1186/s13023-023-02663-z] [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: 02/14/2022] [Accepted: 03/11/2023] [Indexed: 03/30/2023] Open
Abstract
BACKGROUND AND OBJECTIVE The diagnosis of rare diseases (RDs) is often challenging due to their rarity, variability and the high number of individual RDs, resulting in a delay in diagnosis with adverse effects for patients and healthcare systems. The development of computer assisted diagnostic decision support systems could help to improve these problems by supporting differential diagnosis and by prompting physicians to initiate the right diagnostic tests. Towards this end, we developed, trained and tested a machine learning model implemented as part of the software called Pain2D to classify four rare diseases (EDS, GBS, FSHD and PROMM), as well as a control group of unspecific chronic pain, from pen-and-paper pain drawings filled in by patients. METHODS Pain drawings (PDs) were collected from patients suffering from one of the four RDs, or from unspecific chronic pain. The latter PDs were used as an outgroup in order to test how Pain2D handles more common pain causes. A total of 262 (59 EDS, 29 GBS, 35 FSHD, 89 PROMM, 50 unspecific chronic pain) PDs were collected and used to generate disease specific pain profiles. PDs were then classified by Pain2D in a leave-one-out-cross-validation approach. RESULTS Pain2D was able to classify the four rare diseases with an accuracy of 61-77% with its binary classifier. EDS, GBS and FSHD were classified correctly by the Pain2D k-disease classifier with sensitivities between 63 and 86% and specificities between 81 and 89%. For PROMM, the k-disease classifier achieved a sensitivity of 51% and specificity of 90%. CONCLUSIONS Pain2D is a scalable, open-source tool that could potentially be trained for all diseases presenting with pain.
Collapse
Affiliation(s)
- D Emmert
- Center for Rare Diseases Bonn (ZSEB), University Hospital Bonn, Bonn, Germany
- Institute for Virology, University Hospital Bonn, Bonn, Germany
| | - N Szczypien
- Institute for Information Engineering, Ostfalia University of Applied Sciences, Wolfenbüttel, Germany
- Biostatistics Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Tim T A Bender
- Center for Rare Diseases Bonn (ZSEB), University Hospital Bonn, Bonn, Germany
| | - L Grigull
- Center for Rare Diseases Bonn (ZSEB), University Hospital Bonn, Bonn, Germany
| | - A Gass
- Clinic for Anesthesiology and Operative Intensive Care Medicine, Department of Pain Medicine, University Hospital Bonn, Bonn, Germany
| | - C Link
- Clinic for Anesthesiology and Operative Intensive Care Medicine, Department of Pain Medicine, University Hospital Bonn, Bonn, Germany
| | - F Klawonn
- Institute for Information Engineering, Ostfalia University of Applied Sciences, Wolfenbüttel, Germany
- Biostatistics Group, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - R Conrad
- Department of Psychosomatic Medicine and Psychotherapy, University Hospital Muenster, Muenster, Germany.
| | - M Mücke
- Institute for Digitalization and General Medicine, University Hospital RWTH Aachen, Aachen, Germany.
- Center for Rare Diseases Aachen (ZSEA), University Hospital RWTH Aachen, Aachen, Germany.
| | - J Sellin
- Institute for Digitalization and General Medicine, University Hospital RWTH Aachen, Aachen, Germany.
- Center for Rare Diseases Aachen (ZSEA), University Hospital RWTH Aachen, Aachen, Germany.
| |
Collapse
|
8
|
Wamkpah NS, Chi JJ. Facial paresis as the first sign in atypical facioscapulohumeral muscular dystrophy. OTOLARYNGOLOGY CASE REPORTS 2022. [DOI: 10.1016/j.xocr.2022.100468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022] Open
|
9
|
Lu-Nguyen N, Dickson G, Malerba A, Popplewell L. Long-Term Systemic Treatment of a Mouse Model Displaying Chronic FSHD-like Pathology with Antisense Therapeutics That Inhibit DUX4 Expression. Biomedicines 2022; 10:biomedicines10071623. [PMID: 35884928 PMCID: PMC9313434 DOI: 10.3390/biomedicines10071623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/03/2022] [Accepted: 07/05/2022] [Indexed: 11/29/2022] Open
Abstract
Silencing the expression of the double homeobox 4 (DUX4) gene offers great potential for the treatment of facioscapulohumeral muscular dystrophy (FSHD). Several research groups have recently reported promising results using systemic antisense therapy in a transgenic small animal model of FSHD, the ACTA1-MCM/FLExDUX4 mouse model. However, the treatment was applied in non-DUX4-induced mice or shortly after DUX4 activation, which resulted in conditions that do not correctly represent the situation in a clinic. Here, we generated progressive FSHD-like pathology in ACTA1-MCM/FLExDUX4 mice and then treated the animals with vivoPMO-PACS4, an antisense compound that efficiently downregulates DUX4. To best mimic the translation of this treatment in clinical settings, the systemic antisense oligonucleotide administration was delayed to 3 weeks after the DUX4 activation so that the pathology was established at the time of the treatment. The chronic administration of vivoPMO-PACS4 for 8 weeks downregulated the DUX4 expression by 60%. Consequently, the treated mice showed an increase by 18% in body-wide muscle mass and 32% in muscle strength, and a reduction in both myofiber central nucleation and muscle fibrosis by up to 29% and 37%, respectively. Our results in a more suitable model of FSHD pathology confirm the efficacy of vivoPMO-PACS4 administration, and highlight the significant benefit provided by the long-term treatment of the disease.
Collapse
Affiliation(s)
- Ngoc Lu-Nguyen
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham TW20 0EX, UK; (N.L.-N.); (G.D.)
| | - George Dickson
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham TW20 0EX, UK; (N.L.-N.); (G.D.)
| | - Alberto Malerba
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham TW20 0EX, UK; (N.L.-N.); (G.D.)
- Correspondence: (A.M.); (L.P.)
| | - Linda Popplewell
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham TW20 0EX, UK; (N.L.-N.); (G.D.)
- National Horizons Centre, Teesside University, Darlington DL1 1HG, UK
- Correspondence: (A.M.); (L.P.)
| |
Collapse
|
10
|
Wong CJ, Wang L, Holers VM, Frazer-Abel A, van der Maarel SM, Tawil R, Statland JM, Tapscott SJ. Elevated plasma complement components in facioscapulohumeral dystrophy. Hum Mol Genet 2022; 31:1821-1829. [PMID: 34919696 PMCID: PMC9169453 DOI: 10.1093/hmg/ddab364] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Revised: 12/07/2021] [Accepted: 12/14/2021] [Indexed: 11/12/2022] Open
Abstract
Advances in understanding the pathophysiology of facioscapulohumeral dystrophy (FSHD) have led to several therapeutic approaches entering clinical trials and an increased need to develop biomarkers of disease activity and progression. Multiple prior studies have shown early elevation of RNAs encoding components of the complement pathways and relatively widespread activated complement complexes by immunodetection in FSHD muscle. The current study tested plasma from two independent cohorts of FSHD and control subjects and found elevated complement components in both FSHD cohorts. Combining subjects from both cohorts identified complement factors that best distinguished FSHD and controls. Within the FSHD group, a subset of subjects showed elevation in multiple complement components. Together these findings suggest the need for future studies to determine whether measurements of complement activation can be used as a non-invasive measurement of FSHD disease activity, progression and/or response to therapies. In addition, with the ongoing expansion of complement therapeutic approaches, consideration for precision-based targeting of this pathway is appropriate.
Collapse
Affiliation(s)
- Chao-Jen Wong
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | - Leo Wang
- Department of Neurology, University of Washington, Seattle, WA 98105, USA
| | - V Michael Holers
- Division of Rheumatology, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | - Ashley Frazer-Abel
- Exsera BioLabs, Division of Rheumatalogy, University of Colorado School of Medicine, Aurora, CO 80045, USA
| | | | - Rabi Tawil
- Department of Neurology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Jeffrey M Statland
- Department of Neurology, University of Kansas Medical Center, Kansas City, KA 66160, 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
|
11
|
Spatio-temporal gait differences in facioscapulohumeral muscular dystrophy during single and dual task overground walking - A pilot study. J Clin Transl Res 2022; 8:166-175. [PMID: 35734417 PMCID: PMC9206720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 12/29/2021] [Accepted: 01/11/2022] [Indexed: 11/07/2022] Open
Abstract
Background Facioscapulohumeral muscular dystrophy (FSHD) is a rare genetic muscle disorder leading to progressive muscle loss over time. Research indicates that this progressive muscular atrophy can negatively impact spatio-temporal gait characteristics, but this is not always the case during early-onset or mild cases of the disease. In addition, the performance of a secondary task during overground walking may elucidate greater deficits in spatio-temporal characteristics of gait. However, such dual task effects on FSHD gait have not been studied thus far. Aim The current study aimed to (a) quantify changes in spatio-temporal gait parameters in individuals with FSHD using the Tekscan Strideway gait mat system, (b) measure the dual task (DT) effects on cadence and gait velocity during single task (ST) and DT overground walking in FSHD and healthy controls and (c) investigate the correlation between the gait parameters and the methylation status in FSHD. Methods Nine FSHD (M±SD=52.78±14.69 years) and nine nearly matched healthy controls (M±SD=50.11±16.18 years) performed five ST and five DT walking in a pseudo-randomized order. The DT included a serial 7's subtraction task from a random number between 50 and 100. Dependent variables: Cadence (steps/min) and gait velocity (cm/sec) were obtained from Tekscan Strideway (30 Hz, Boston, MA). Results The pairwise comparison indicated that cadence was significantly different for both ST (P<0.004) and DT (P<0.02) where FSHD showed lower cadence compared to controls. Gait velocity was also significantly lower for FSHD during ST (P<0.004) and DT (P<0.008). Multilevel modeling (MLM) approach revealed a group by task interaction for cadence (P<0.05) and gait velocity (P<0.001). The interaction showed a significant difference between ST and DT in controls for cadence and gait velocity. However, there was no difference between ST and DT in FSHD. Finally, a comparison of methylation percentage versus gait parameters revealed a significant negative correlation coefficient for cadence but not for gait velocity. Conclusion These results indicate specific pairwise differences in both ST and DT walking, observed in the gait parameters as decreased cadence and gait velocity during ST and DT. In addition, the MLM showed that controls exhibited the DT cost as expected but FSHD did not for cadence and gait velocity. Relevance for Patients ST appears to be sufficiently challenging in FSHD and results in overall declines in spatio-temporal characteristics of gait. Further research is needed to test this paradigm with early-onset or mild cases to track disease progression and its effects on ambulation.
Collapse
|
12
|
Triantafyllidou O, Stavridis K, Kastora SL, Vlahos N. Road to conception and successful delivery for a facioscapulohumeral muscular dystrophy patient. SAGE Open Med Case Rep 2022; 10:2050313X221081359. [PMID: 35251661 PMCID: PMC8891829 DOI: 10.1177/2050313x221081359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/30/2022] [Indexed: 11/16/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy is a muscular dystrophy affecting all ages, primarily people in the second decade. The disease is initially presented with face, shoulder girdle, and upper arm involvement, followed by lower extremity muscle weakness. Disease progression is usually slow, although about one-fifth of patients will require a wheelchair to accommodate mobility. Women with this muscular dystrophy could rarely have poor birth outcomes, with facioscapulohumeral muscular dystrophy symptom deterioration post-partum. In this study, we present a case of a woman with a genetically confirmed facioscapulohumeral muscular dystrophy 1 who underwent cesarean section with epidural anesthesia with favorable outcomes following the procedure. Eight months post cesarean section, the patient reported no facioscapulohumeral muscular dystrophy symptom deterioration. We reviewed the literature with emphasis on large studies concerning facioscapulohumeral muscular dystrophy and birth outcomes and concluded that the hereby presented approach is important for the comprehensive obstetric care and future risk assessment and management in such patients.
Collapse
Affiliation(s)
- Olga Triantafyllidou
- 2nd Department of Obstetrics and Gynaecology, Aretaieion Hospital, University of Athens, Athens, Greece
| | - Konstantinos Stavridis
- 2nd Department of Obstetrics and Gynaecology, Aretaieion Hospital, University of Athens, Athens, Greece
| | - Stavroula Lila Kastora
- Acute Medicine, Grampian University Hospitals NHS Trust, Aberdeen, UK
- School of Medicine, University of Aberdeen, Aberdeen, UK
| | - Nikolaos Vlahos
- 2nd Department of Obstetrics and Gynaecology, Aretaieion Hospital, University of Athens, Athens, Greece
| |
Collapse
|
13
|
Santos VBD, Saute JAM, Jacinto-Scudeiro LA, Ayres A, Rech RS, Oliveira AAD, Olchik MR. Speech and swallowing characteristics in patients with facioscapulohumeral muscular dystrophy. ARQUIVOS DE NEURO-PSIQUIATRIA 2022; 80:368-374. [PMID: 35195226 DOI: 10.1590/0004-282x-anp-2021-0034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 07/31/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Although facial muscle weakness is common in patients with Facioscapulohumeral Muscular Dystrophy (FSHD), the literature is scarce on the speech and swallowing aspects. OBJECTIVE To investigate speech and swallowing patterns in FSHD and assess the correlation with clinical data. METHODS A cross-sectional study was conducted. Patients with clinical confirmation of FSHD and aged above 18 years were included and paired with healthy control individuals by age and gender. Individuals who had neurological conditions that could interfere with test results were excluded. The following assessments were applied: speech tests (acoustic and auditory-perceptual analysis); swallowing tests with the Northwestern Dysphagia Patient Check Sheet (NDPCS), the Eat Assessment Tool (EAT-10), the Speech Therapy Protocol for Dysphagia Risk (PARD), and the Functional Oral Intake Scale (FOIS); disease staging using the modified Gardner-Medwin-Walton scale (GMWS); and quality of life with the Medical Outcomes Study 36-Item Short-Form Health Survey (SF-36). The correlation between test results and clinical data was verified by non-parametric statistics. RESULTS Thirteen individuals with FSHD and 10 healthy controls were evaluated. The groups presented significant differences in the motor bases of phonation and breathing. Regarding swallowing, two (15%) individuals presented mild dysphagia and seven (53.8%) showed reduced facial muscles strength. These results were not correlated with duration of the disease, age at symptoms onset, and quality of life. Dysphagia was related to worsening disease severity. CONCLUSIONS FSHD patients presented mild dysarthria and dysphagia. Frequent monitoring of these symptoms could be an important way to provide early rehabilitation and better quality of life.
Collapse
Affiliation(s)
- Vanessa Brzoskowski Dos Santos
- Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Ciências da Reabilitação, Porto Alegre RS, Brazil
| | - Jonas Alex Morales Saute
- Universidade Federal do Rio Grande do Sul, Departamento de Medicina Interna, Porto Alegre RS, Brazil
| | | | - Annelise Ayres
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre RS, Brazil
| | - Rafaela Soares Rech
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre RS, Brazil
| | - Alcyr Alves de Oliveira
- Universidade Federal de Ciências da Saúde de Porto Alegre, Programa de Pós-Graduação em Ciências da Reabilitação, Porto Alegre RS, Brazil
| | - Maira Rozenfeld Olchik
- Universidade Federal do Rio Grande do Sul, Departamento de Cirurgia e Ortopedia, Porto Alegre RS, Brazil
| |
Collapse
|
14
|
Lu-Nguyen N, Malerba A, Antoni Pineda M, Dickson G, Popplewell LJ. Improving molecular and histopathology in diaphragm muscle of the double transgenic ACTA1-MCM/FLExDUX4 mouse model of FSHD with systemic antisense therapy. Hum Gene Ther 2022; 33:923-935. [PMID: 35078334 DOI: 10.1089/hum.2021.251] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a rare muscle dystrophy causing muscle weakness initially in the face, shoulders and upper arms, and extended to lower body muscles as the disease progresses. Respiratory restriction in FSHD is increasingly reported to be more common and severe than previously thought, with the involvement of diaphragm weakness in pulmonary insufficiency being under debate. As aberrant expression of the double homeobox 4 (DUX4) gene is the prime cause of FSHD, we and others have developed numerous strategies and reported promising results on downregulating DUX4 expression in both cellular and animal models of FSHD. However, the effect of DUX4 and anti-DUX4 approaches on diaphragm muscle has not been elucidated. Here we show that toxic DUX4 expression causes pathology that affects the diaphragm of ACTA1-MCM/FLExDUX4 mouse model of FSHD at both molecular and histological levels. Of importance, a systemic antisense treatment that suppresses DUX4 and target genes expression by 50% significantly improves muscle regeneration and muscle fibrosis, and prevents modification in myofiber type composition, supporting its development as a treatment for FSHD.
Collapse
Affiliation(s)
- Ngoc Lu-Nguyen
- Royal Holloway University of London, 3162, Department of Biological Sciences, Egham, Surrey, United Kingdom of Great Britain and Northern Ireland;
| | - Alberto Malerba
- Royal Holloway University of London, 3162, Department of Biological Sciences, Egham, Surrey, United Kingdom of Great Britain and Northern Ireland;
| | - Marina Antoni Pineda
- Royal Holloway University of London, 3162, Department of Biological Sciences, Egham, Surrey, United Kingdom of Great Britain and Northern Ireland;
| | - George Dickson
- Royal Holloway University of London, 3162, Department of Biological Sciences, Egham, Surrey, United Kingdom of Great Britain and Northern Ireland;
| | - Linda J Popplewell
- Royal Holloway University of London, 3162, Department of Biological Sciences, Egham, Surrey, United Kingdom of Great Britain and Northern Ireland;
| |
Collapse
|
15
|
Santos VBD, Saute JAM, Jacinto-Scudeiro LA, Ayres A, Rech RS, Oliveira AAD, Olchik MR. Cognitive profile of patients with facioscapulohumeral muscular dystrophy. Dement Neuropsychol 2021; 15:541-547. [PMID: 35509802 PMCID: PMC9018092 DOI: 10.1590/1980-57642021dn15-040015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 07/13/2021] [Indexed: 08/30/2023] Open
Abstract
Although it is predominantly a muscular disease, impairments in the central nervous system in patients with facioscapulohumeral muscular dystrophy (FSHD) have been described in the literature.
Collapse
Affiliation(s)
| | - Jonas Alex Morales Saute
- Department of Internal Medicine, Brazil; Postgraduate Program in Medicine: Medical Sciences, Brazil
| | | | | | | | | | - Maira Rozenfeld Olchik
- Postgraduate Program in Medicine: Medical Sciences, Brazil; Department of Surgery and Orthopedics, Brazil
| |
Collapse
|
16
|
Yedigaryan L, Sampaolesi M. Therapeutic Implications of miRNAs for Muscle-Wasting Conditions. Cells 2021; 10:cells10113035. [PMID: 34831256 PMCID: PMC8616481 DOI: 10.3390/cells10113035] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 10/28/2021] [Accepted: 10/30/2021] [Indexed: 12/14/2022] Open
Abstract
MicroRNAs (miRNAs) are small, non-coding RNA molecules that are mainly involved in translational repression by binding to specific messenger RNAs. Recently, miRNAs have emerged as biomarkers, relevant for a multitude of pathophysiological conditions, and cells can selectively sort miRNAs into extracellular vesicles for paracrine and endocrine effects. In the overall context of muscle-wasting conditions, a multitude of miRNAs has been implied as being responsible for the typical dysregulation of anabolic and catabolic pathways. In general, chronic muscle disorders are associated with the main characteristic of a substantial loss in muscle mass. Muscular dystrophies (MDs) are a group of genetic diseases that cause muscle weakness and degeneration. Typically, MDs are caused by mutations in those genes responsible for upholding the integrity of muscle structure and function. Recently, the dysregulation of miRNA levels in such pathological conditions has been reported. This revelation is imperative for both MDs and other muscle-wasting conditions, such as sarcopenia and cancer cachexia. The expression levels of miRNAs have immense potential for use as potential diagnostic, prognostic and therapeutic biomarkers. Understanding the role of miRNAs in muscle-wasting conditions may lead to the development of novel strategies for the improvement of patient management.
Collapse
Affiliation(s)
- Laura Yedigaryan
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium;
| | - Maurilio Sampaolesi
- Translational Cardiomyology Laboratory, Stem Cell Biology and Embryology, Department of Development and Regeneration, KU Leuven, 3000 Leuven, Belgium;
- Histology and Medical Embryology Unit, Department of Anatomy, Histology, Forensic Medicine and Orthopedics, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence:
| |
Collapse
|
17
|
Lu-Nguyen N, Malerba A, Herath S, Dickson G, Popplewell L. Systemic antisense therapeutics inhibiting DUX4 expression ameliorates FSHD-like pathology in an FSHD mouse model. Hum Mol Genet 2021; 30:1398-1412. [PMID: 33987655 PMCID: PMC8283208 DOI: 10.1093/hmg/ddab136] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 05/03/2021] [Accepted: 05/04/2021] [Indexed: 12/11/2022] Open
Abstract
Aberrant expression of the double homeobox 4 (DUX4) gene in skeletal muscle causes muscle deterioration and weakness in Facioscapulohumeral muscular dystrophy (FSHD). Since the presence of a permissive pLAM1 polyadenylation signal is essential for stabilization of DUX4 mRNA and translation of DUX4 protein, disrupting the function of this structure can prevent expression of DUX4. We and others have shown promising results using antisense approaches to reduce DUX4 expression in vitro and in vivo following local intramuscular administration. Here we demonstrate that further development of the antisense chemistries enhances in vitro antisense efficacy. The optimal chemistry was conjugated to a cell-penetrating moiety and was systemically administered into the tamoxifen-inducible Cre-driver FLExDUX4 double-transgenic mouse model of FSHD. After four weekly treatments, mRNA quantities of DUX4 and target genes were reduced by 50% that led to 12% amelioration in muscle atrophy, 52% improvement in in situ muscle strength, 17% reduction in muscle fibrosis and prevention of shift in the myofiber type profile. Systemic DUX4 inhibition also significantly improved the locomotor activity and reduced the fatigue level by 22%. Our data demonstrate that the optimized antisense approach has potential of being further developed as a therapeutic strategy for FSHD.
Collapse
Affiliation(s)
- Ngoc Lu-Nguyen
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Alberto Malerba
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Shan Herath
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - George Dickson
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| | - Linda Popplewell
- Department of Biological Sciences, School of Life Sciences and the Environment, Royal Holloway University of London, Egham, Surrey TW20 0EX, UK
| |
Collapse
|
18
|
Abstract
Increased understanding of disease pathophysiology and advances in gene therapies and drug technologies are revolutionizing treatment of muscular dystrophies and motor neuron disorders (MNDs). New drugs have been approved for Duchenne muscular dystrophy, spinal muscular atrophy, and amyotrophic lateral sclerosis. For other diseases, new targets have been identified, and new therapies are in clinical trials. The impact of such therapies will be fully understood only in the next decades. Cost burden and accessibility are major challenges in the wide application of new drugs. This article reviews advances in gene therapies, newly approved drugs, and therapeutic promises in muscular dystrophies and MNDs.
Collapse
|
19
|
Bourguiba R, Masingue M, Marc PJ, Lenglet T, Bachmeyer C. A Great Athlete with Muscular Weakness. Am J Med 2021; 134:e109-e110. [PMID: 32835688 DOI: 10.1016/j.amjmed.2020.07.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/23/2020] [Accepted: 07/24/2020] [Indexed: 11/18/2022]
Affiliation(s)
- Rim Bourguiba
- Department of Internal Medicine, Assistance Publique-Hôpitaux de Paris, Hopital Tenon, Paris, France.
| | - Marion Masingue
- Reference Center for Neuromuscular Diseases Nord-Est-Ile de France, Institute of Myology, CHU Pitié Salpetrière, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Pierre-Jean Marc
- Department of Genetics, Hôpital Cochin, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Thimothée Lenglet
- Department of Clinical Neurophysiology, Hôpitaux Universitaires Pitié-Salpêtrière Charles Foix, Assistance Publique-Hôpitaux de Paris (APHP), Paris, France
| | - Claude Bachmeyer
- Department of Internal Medicine, Assistance Publique-Hôpitaux de Paris, Hopital Tenon, Paris, France
| |
Collapse
|
20
|
A team approach in the diagnosis and management of facioscapulohumeral muscular dystrophy: a case report. JSES Int 2020; 4:485-490. [PMID: 32939472 PMCID: PMC7479020 DOI: 10.1016/j.jseint.2020.04.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
|
21
|
Chaudhari N, Rickard AM, Roy S, Dröge P, Makhija H. A non-viral genome editing platform for site-specific insertion of large transgenes. Stem Cell Res Ther 2020; 11:380. [PMID: 32883366 PMCID: PMC7650303 DOI: 10.1186/s13287-020-01890-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/22/2020] [Accepted: 08/18/2020] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The precise, functional and safe insertion of large DNA payloads into host genomes offers versatility in downstream genetic engineering-associated applications, spanning cell and gene therapies, therapeutic protein production, high-throughput cell-based drug screening and reporter cell lines amongst others. Employing viral- and non-viral-based genome engineering tools to achieve specific insertion of large DNA-despite being successful in E. coli and animal models-still pose challenges in the human system. In this study, we demonstrate the applicability of our lambda integrase-based genome insertion tool for human cell and gene therapy applications that require insertions of large functional genes, as exemplified by the integration of a functional copy of the F8 gene and a Double Homeobox Protein 4 (DUX4)-based reporter cassette for potential hemophilia A gene therapy and facioscapulohumeral muscular dystrophy (FSHD)-based high-throughput drug screening purposes, respectively. Thus, we present a non-viral genome insertion tool for safe and functional delivery of large seamless DNA cargo into the human genome that can enable novel designer cell-based therapies. METHODS Previously, we have demonstrated the utility of our phage λ-integrase platform to generate seamless vectors and subsequently achieve functional integration of large-sized DNA payloads at defined loci in the human genome. To further explore this tool for therapeutic applications, we used pluripotent human embryonic stem cells (hESCs) to integrate large seamless vectors comprising a 'gene of interest'. Clonal cell populations were screened for the correct integration events and further characterized by southern blotting, gene expression and protein activity assays. In the case of our hemophilia A-related study, clones were differentiated to confirm that the targeted locus is active after differentiation and actively express and secrete Factor VIII. RESULTS The two independent approaches demonstrated specific and functional insertions of a full-length blood clotting F8 expression cassette of ~ 10 kb and of a DUX4 reporter cassette of ~ 7 kb in hESCs. CONCLUSION We present a versatile tool for site-specific human genome engineering with large transgenes for cell/gene therapies and other synthetic biology and biomedical applications.
Collapse
Affiliation(s)
- Namrata Chaudhari
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Republic of Singapore
| | - Amanda M Rickard
- Genea Biocells, 11099 North Torrey Pines Road, Suite 210, La Jolla, CA, 92037, USA
| | - Suki Roy
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Republic of Singapore
| | - Peter Dröge
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Republic of Singapore.
| | - Harshyaa Makhija
- School of Biological Sciences, Nanyang Technological University, Singapore, 637551, Republic of Singapore.
| |
Collapse
|
22
|
DNA methylation in satellite repeats disorders. Essays Biochem 2020; 63:757-771. [PMID: 31387943 DOI: 10.1042/ebc20190028] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 07/22/2019] [Accepted: 07/24/2019] [Indexed: 02/06/2023]
Abstract
Despite the tremendous progress made in recent years in assembling the human genome, tandemly repeated DNA elements remain poorly characterized. These sequences account for the vast majority of methylated sites in the human genome and their methylated state is necessary for this repetitive DNA to function properly and to maintain genome integrity. Furthermore, recent advances highlight the emerging role of these sequences in regulating the functions of the human genome and its variability during evolution, among individuals, or in disease susceptibility. In addition, a number of inherited rare diseases are directly linked to the alteration of some of these repetitive DNA sequences, either through changes in the organization or size of the tandem repeat arrays or through mutations in genes encoding chromatin modifiers involved in the epigenetic regulation of these elements. Although largely overlooked so far in the functional annotation of the human genome, satellite elements play key roles in its architectural and topological organization. This includes functions as boundary elements delimitating functional domains or assembly of repressive nuclear compartments, with local or distal impact on gene expression. Thus, the consideration of satellite repeats organization and their associated epigenetic landmarks, including DNA methylation (DNAme), will become unavoidable in the near future to fully decipher human phenotypes and associated diseases.
Collapse
|
23
|
Lim KRQ, Nguyen Q, Yokota T. DUX4 Signalling in the Pathogenesis of Facioscapulohumeral Muscular Dystrophy. Int J Mol Sci 2020; 21:E729. [PMID: 31979100 PMCID: PMC7037115 DOI: 10.3390/ijms21030729] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 01/17/2020] [Accepted: 01/18/2020] [Indexed: 12/17/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is a disabling inherited muscular disorder characterized by asymmetric, progressive muscle weakness and degeneration. Patients display widely variable disease onset and severity, and sometimes present with extra-muscular symptoms. There is a consensus that FSHD is caused by the aberrant production of the double homeobox protein 4 (DUX4) transcription factor in skeletal muscle. DUX4 is normally expressed during early embryonic development, and is then effectively silenced in all tissues except the testis and thymus. Its reactivation in skeletal muscle disrupts numerous signalling pathways that mostly converge on cell death. Here, we review studies on DUX4-affected pathways in skeletal muscle and provide insights into how understanding these could help explain the unique pathogenesis of FSHD.
Collapse
Affiliation(s)
- Kenji Rowel Q. Lim
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (Q.N.)
| | - Quynh Nguyen
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (Q.N.)
| | - Toshifumi Yokota
- Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB T6G2H7, Canada; (K.R.Q.L.); (Q.N.)
- The Friends of Garrett Cumming Research & Muscular Dystrophy Canada, HM Toupin Neurological Science Research Chair, Edmonton, AB T6G2H7, Canada
| |
Collapse
|
24
|
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: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
25
|
Amini Chermahini G, Rashnonejad A, Harper SQ. RNAscope in situ hybridization-based method for detecting DUX4 RNA expression in vitro. RNA (NEW YORK, N.Y.) 2019; 25:1211-1217. [PMID: 31209064 PMCID: PMC6800509 DOI: 10.1261/rna.070177.118] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 06/07/2019] [Indexed: 06/09/2023]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is among the most common forms of muscular dystrophy. FSHD is caused by aberrant expression of the toxic DUX4 gene in muscle. Detecting endogenous DUX4 in patient tissue using conventional methods can be challenging, due to the low level of DUX4 expression. Therefore, developing simple and trustworthy DUX4 detection methods is an important need in the FSHD field. Here, we describe such a method, which uses the RNAscope assay, an RNA in situ hybridization (ISH) technology. We show that a custom-designed RNAscope assay can detect overexpressed DUX4 mRNA in transfected HEK293 cells and endogenous DUX4 mRNA in FSHD patient-derived myotubes. The RNAscope assay was highly sensitive for tracking reductions in DUX4 mRNA following treatment with our therapeutic mi405 microRNA, suggesting that RNAscope-based DUX4 expression assays could be developed as a prospective outcome measure in therapy trials. This study could set the stage for optimizing and developing a new, rapid RNA ISH-based molecular diagnostic assay for future clinical use in the FSHD field.
Collapse
Affiliation(s)
| | - Afrooz Rashnonejad
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205, USA
| | - Scott Q Harper
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio 43205, USA
- Department of Pediatrics, The Ohio State University, Columbus, Ohio 43205, USA
| |
Collapse
|
26
|
Hamel J, Johnson N, Tawil R, Martens WB, Dilek N, McDermott MP, Heatwole C. Patient-Reported Symptoms in Facioscapulohumeral Muscular Dystrophy (PRISM-FSHD). Neurology 2019; 93:e1180-e1192. [PMID: 31409737 DOI: 10.1212/wnl.0000000000008123] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 04/24/2019] [Indexed: 01/23/2023] Open
Abstract
OBJECTIVE To determine the frequency and relative importance of the most meaningful symptoms in facioscapulohumeral muscular dystrophy (FSHD) and to identify the demographic and clinical features that are associated with the greatest disease burden in this population. METHODS We performed a cross-sectional study involving 328 participants with FSHD. Collectively, participants reported the prevalence and relative importance of 274 symptoms and 15 symptomatic themes. We assessed the association between symptomatic theme prevalence and participants' age, sex, disease duration, pain level, employment status, and education. RESULTS Participants answered >48,000 questions regarding their disease burden. The symptomatic themes with the highest prevalence in our sample were problems with shoulders or arms (96.9%), limitations with activities (94.7%), core weakness (93.8%), fatigue (93.8%), limitations with mobility and walking (93.6%), changed body image due to the disease (91.6%), and pain (87.7%). Problems with shoulders and arms and limitations with mobility and walking had the greatest effect on participants' lives. Employment status and the report of pain had the most extensive association with the prevalence of symptoms, with employment being associated with 8 of 15 of the symptomatic themes and pain being associated with 7 of 15 of the symptomatic themes. Men and women with FSHD experienced a similar prevalence of all symptomatic themes. CONCLUSIONS Adults with FSHD experience a variety of symptoms that play an important role in their disease burden. These symptoms have a variable prevalence and importance in the FSHD population and are associated with disease duration, employment status, and pain level.
Collapse
Affiliation(s)
- Johanna Hamel
- From the Department of Neurology (J.H., R.T., W.B.M., N.D., M.P.M., C.H.) and Department of Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; and Department of Neurology (N.J.), Virginia Commonwealth University, Richmond.
| | - Nicholas Johnson
- From the Department of Neurology (J.H., R.T., W.B.M., N.D., M.P.M., C.H.) and Department of Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; and Department of Neurology (N.J.), Virginia Commonwealth University, Richmond
| | - Rabi Tawil
- From the Department of Neurology (J.H., R.T., W.B.M., N.D., M.P.M., C.H.) and Department of Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; and Department of Neurology (N.J.), Virginia Commonwealth University, Richmond
| | - William B Martens
- From the Department of Neurology (J.H., R.T., W.B.M., N.D., M.P.M., C.H.) and Department of Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; and Department of Neurology (N.J.), Virginia Commonwealth University, Richmond
| | - Nuran Dilek
- From the Department of Neurology (J.H., R.T., W.B.M., N.D., M.P.M., C.H.) and Department of Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; and Department of Neurology (N.J.), Virginia Commonwealth University, Richmond
| | - Michael P McDermott
- From the Department of Neurology (J.H., R.T., W.B.M., N.D., M.P.M., C.H.) and Department of Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; and Department of Neurology (N.J.), Virginia Commonwealth University, Richmond
| | - Chad Heatwole
- From the Department of Neurology (J.H., R.T., W.B.M., N.D., M.P.M., C.H.) and Department of Biostatistics and Computational Biology (M.P.M.), University of Rochester Medical Center, NY; and Department of Neurology (N.J.), Virginia Commonwealth University, Richmond
| |
Collapse
|
27
|
Lemmers RJLF, van der Vliet PJ, Vreijling JP, Henderson D, van der Stoep N, Voermans N, van Engelen B, Baas F, Sacconi S, Tawil R, van der Maarel SM. Cis D4Z4 repeat duplications associated with facioscapulohumeral muscular dystrophy type 2. Hum Mol Genet 2019; 27:3488-3497. [PMID: 30281091 DOI: 10.1093/hmg/ddy236] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 06/15/2018] [Indexed: 12/26/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy, known in genetic forms FSHD1 and FSHD2, is associated with D4Z4 repeat array chromatin relaxation and somatic derepression of DUX4 located in D4Z4. A complete copy of DUX4 is present on 4qA chromosomes, but not on the D4Z4-like repeats of chromosomes 4qB or 10. Normally, the D4Z4 repeat varies between 8 and 100 units, while in FSHD1 it is only 1-10 units. In the rare genetic form FSHD2, a combination of a 4qA allele with a D4Z4 repeat size of 8-20 units and heterozygous pathogenic variants in the chromatin modifier SMCHD1 causes DUX4 derepression and disease. In this study, we identified 11/79 (14%) FSHD2 patients with unusually large 4qA alleles of 21-70 D4Z4 units. By a combination of Southern blotting and molecular combing, we show that 8/11 (73%) of these unusually large 4qA alleles represent duplication alleles in which the long D4Z4 repeat arrays are followed by a small FSHD-sized D4Z4 repeat array duplication. We also show that these duplication alleles are associated with DUX4 expression. This duplication allele frequency is significantly higher than in controls (2.9%), FSHD1 patients (1.4%) and in FSHD2 patients with typical 4qA alleles of 8-20 D4Z4 units (1.5%). Segregation analysis shows that, similar to typical 8-20 units FSHD2 alleles, duplication alleles only cause FSHD in combination with a pathogenic variant in SMCHD1. We conclude that cis duplications of D4Z4 repeats explain DUX4 expression and disease presentation in FSHD2 families with unusual long D4Z4 repeats on 4qA chromosomes.
Collapse
Affiliation(s)
| | | | - Jeroen P Vreijling
- Laboratory for Diagnostic Genome Analysis, Leiden University Medical Center, Leiden, RC, Netherlands
| | - Don Henderson
- Neuromuscular Disease Unit, Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Nienke van der Stoep
- Laboratory for Diagnostic Genome Analysis, Leiden University Medical Center, Leiden, RC, Netherlands
| | - Nicol Voermans
- Neuromuscular Centre Nijmegen, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, HB, Netherlands
| | - Baziel van Engelen
- Neuromuscular Centre Nijmegen, Department of Neurology, Donders Institute for Brain, Cognition and Behaviour, Radboud University Medical Center, Nijmegen, HB, Netherlands
| | - Frank Baas
- Laboratory for Diagnostic Genome Analysis, Leiden University Medical Center, Leiden, RC, Netherlands
| | - Sabrina Sacconi
- Centre de Référence des Maladies Neuromusculaires and CNRS UMR6543, Nice University Hospital, Nice, France
| | - Rabi Tawil
- Neuromuscular Disease Unit, Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | | |
Collapse
|
28
|
Runte M, Spiesshoefer J, Heidbreder A, Dreher M, Young P, Brix T, Boentert M. Sleep-related breathing disorders in facioscapulohumeral dystrophy. Sleep Breath 2019; 23:899-906. [PMID: 31025273 DOI: 10.1007/s11325-019-01843-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 04/04/2019] [Accepted: 04/08/2019] [Indexed: 11/28/2022]
Abstract
PURPOSE Severe manifestations of facioscapulohumeral dystrophy (FSHD) may be associated with sleep-disordered breathing (SDB), including obstructive sleep apnea (OSA) and nocturnal hypoventilation (NH), but prevalence data are scarce. In patients with respiratory muscle weakness, detection of NH can be facilitated by transcutaneous capnometry, but respective data derived from FSHD patients have not yet been published. METHODS We collected sleep studies and capnometry recordings from 31 adult patients with genetically confirmed FSHD who were admitted to our sleep laboratory for first-ever evaluation of sleep-related breathing. Indications for admission included non-restorative sleep, morning headache, or excessive daytime sleepiness. In addition, sleep studies were initiated if symptoms or signs of respiratory muscle weakness were present. Thirty-one subjects with insomnia served as controls for comparison of respiratory measures during sleep. RESULTS In the FSHD group, 17/31 (55%) patients showed OSA and 8 (26%) had NH. NH would have been missed in 7/8 patients if only oximetry criteria of hypoventilation had been applied. Capnography results were correlated with disease severity as reflected by the Clinical Severity Score (CSS). Non-invasive ventilation (NIV) was started in 6 patients with NH and 3 individuals with OSA. Nocturnal continuous positive airway pressure was administered to 2 patients, and positional therapy was sufficient in 4 individuals. In patients initiated on NIV, nocturnal gas exchange already improved in the first night of treatment. CONCLUSIONS SDB is common in adult patients with FSHD complaining of sleep-related symptoms. It may comprise OSA, NH, and most often, the combination of both. Sleep-related hypercapnia is associated with disease severity. Transcutaneous capnometry is superior to pulse oximetry for detection of NH.
Collapse
Affiliation(s)
- Maya Runte
- Department of Neurology, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany
| | - Jens Spiesshoefer
- Department of Neurology, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany
| | - Anna Heidbreder
- Department of Neurology, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany
| | - Michael Dreher
- Department of Pneumology and Intensive Care Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Peter Young
- Medical Park Klinik Reithofpark, Neurology, Bad Feilnbach, Germany
| | - Tobias Brix
- Institute of Medical Informatics, University of Muenster, Muenster, Germany
| | - Matthias Boentert
- Department of Neurology, University Hospital Muenster, Albert-Schweitzer-Campus 1, Building A1, 48149, Muenster, Germany.
| |
Collapse
|
29
|
Huml RA, Uspenskaya-Cadoz O, Dawson J, Slifer Z. Updating the Clinical Picture of Facioscapulohumeral Muscular Dystrophy: Ramifications for Drug Development With Potential Solutions. Ther Innov Regul Sci 2019. [DOI: 10.1177/2168479018820313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
| | | | | | - Zachary Slifer
- North Carolina State University’s College of Veterinary Medicine, Raleigh, North Carolina, USA
| |
Collapse
|
30
|
Giesige CR, Wallace LM, Heller KN, Eidahl JO, Saad NY, Fowler AM, Pyne NK, Al-Kharsan M, Rashnonejad A, Chermahini GA, Domire JS, Mukweyi D, Garwick-Coppens SE, Guckes SM, McLaughlin KJ, Meyer K, Rodino-Klapac LR, Harper SQ. AAV-mediated follistatin gene therapy improves functional outcomes in the TIC-DUX4 mouse model of FSHD. JCI Insight 2018; 3:123538. [PMID: 30429376 DOI: 10.1172/jci.insight.123538] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 10/10/2018] [Indexed: 01/08/2023] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant or digenic disorder linked to derepression of the toxic DUX4 gene in muscle. There is currently no pharmacological treatment. The emergence of DUX4 enabled development of cell and animal models that could be used for basic and translational research. Since DUX4 is toxic, animal model development has been challenging, but progress has been made, revealing that tight regulation of DUX4 expression is critical for creating viable animals that develop myopathy. Here, we report such a model - the tamoxifen-inducible FSHD mouse model called TIC-DUX4. Uninduced animals are viable, born in Mendelian ratios, and overtly indistinguishable from WT animals. Induced animals display significant DUX4-dependent myopathic phenotypes at the molecular, histological, and functional levels. To demonstrate the utility of TIC-DUX4 mice for therapeutic development, we tested a gene therapy approach aimed at improving muscle strength in DUX4-expressing muscles using adeno-associated virus serotype 1.Follistatin (AAV1.Follistatin), a natural myostatin antagonist. This strategy was not designed to modulate DUX4 but could offer a mechanism to improve muscle weakness caused by DUX4-induced damage. AAV1.Follistatin significantly increased TIC-DUX4 muscle mass and strength even in the presence of DUX4 expression, suggesting that myostatin inhibition may be a promising approach to treat FSHD-associated weakness. We conclude that TIC-DUX4 mice are a relevant model to study DUX4 toxicity and, importantly, are useful in therapeutic development studies for FSHD.
Collapse
Affiliation(s)
- Carlee R Giesige
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio, USA.,Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Lindsay M Wallace
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Kristin N Heller
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Jocelyn O Eidahl
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Nizar Y Saad
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Allison M Fowler
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Nettie K Pyne
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Mustafa Al-Kharsan
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Afrooz Rashnonejad
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | | | - Jacqueline S Domire
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Diana Mukweyi
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Sara E Garwick-Coppens
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Susan M Guckes
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - K John McLaughlin
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Kathrin Meyer
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Louise R Rodino-Klapac
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| | - Scott Q Harper
- Biomedical Sciences Graduate Program, The Ohio State University, Columbus, Ohio, USA.,Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, Ohio, USA.,Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
31
|
Li L, Fu F, Li R, Liu Z, Liao C. Prenatal diagnosis and pregnancy outcome analysis of thickened nuchal fold in the second trimester. Medicine (Baltimore) 2018; 97:e13334. [PMID: 30431620 PMCID: PMC6257687 DOI: 10.1097/md.0000000000013334] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
To summarize the results of prenatal diagnoses and pregnancy outcomes of fetuses with thickened nuchal fold (TNF) in the second trimester.From 2009 to 2016, we studied 72 pregnant women with fetal nuchal fold (NF) measurements over 5 mm at 14 to 19 + 6 weeks or 6 mm at 20 to 28 weeks of gestation who received prenatal diagnosis. Karyotypes were first used to detect common chromosomal diseases, and then chromosome microarray analysis (CMA) was performed if karyotypes were normal. Prognoses were followed up by documentation in the hospital or over the telephone.In total, 12 fetuses with chromosomal defects, including 5 pathogenic copy number variants (CNVs) were detected. The risk of chromosomal defects when a TNF was associated with structural malformations (SMs) (35.5%) was much greater than that of an isolated TNF (3.7%) and a TNF associated with soft markers (0%). The rate of SMs when the NF measured ≥10 mm was greater than that NF measured 5 to 7.9 mm or 8 to 9.9 mm. Totally 27 fetuses had adverse pregnancy outcome.A TNF is not only associated with a high risk of trisomy 21 but also with other chromosomal abnormalities, including pathogenic CNVs. The rates of SMs and adverse outcomes increase when the NF thickness increases.
Collapse
|
32
|
A case of facioscapulohumeral muscular dystrophy and myasthenia gravis with positivity of anti-Ach receptor antibody: a fortuitous association? Neurol Sci 2018; 40:195-197. [PMID: 30215155 DOI: 10.1007/s10072-018-3554-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 09/04/2018] [Indexed: 10/28/2022]
|
33
|
Mul K, Heatwole C, Eichinger K, Dilek N, Martens WB, Van Engelen BGM, Tawil R, Statland JM. Electrical impedance myography in facioscapulohumeral muscular dystrophy: A 1-year follow-up study. Muscle Nerve 2018; 58:213-218. [PMID: 29543984 DOI: 10.1002/mus.26127] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/13/2018] [Indexed: 12/14/2022]
Abstract
INTRODUCTION Electrical impedance myography (EIM) is a noninvasive technique for measuring muscle composition and a potential physiological biomarker for facioscapulohumeral muscular dystrophy (FSHD). METHODS Thirty-two participants with genetically confirmed and clinically affected FSHD underwent EIM in 7 muscles bilaterally. Correlations between EIM and baseline clinical measures were used to select EIM variables of interest in FSHD, and EIM and clinical measures were followed for 1 year. RESULTS There were no significant changes in the EIM variables. Although 50-kHZ reactance correlated the strongest with clinical measures at baseline, the 50-211-kHZ phase ratio demonstrated lower within-subject 12-month variability, potentially offering sample size savings for FSHD clinical trial planning. DISCUSSION EIM did not identify significant disease progression over 12 months. It is currently unclear whether this is because of limitations of the technology or the slow rate of disease progression in this cohort of FSHD patients over this period of time. Muscle Nerve 58: 213-218, 2018.
Collapse
Affiliation(s)
- Karlien Mul
- Department of Neurology, University of Kansas Medical Center, 4330 Shawnee Mission Parkway, Suite 323 Fairway, Kansas, 66205, USA.,Department of Neurology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Chad Heatwole
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Katy Eichinger
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Nuran Dilek
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - William B Martens
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Baziel G M Van Engelen
- Department of Neurology, Donders Institute for Brain, Cognition, and Behaviour, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Rabi Tawil
- Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| | - Jeffrey M Statland
- Department of Neurology, University of Kansas Medical Center, 4330 Shawnee Mission Parkway, Suite 323 Fairway, Kansas, 66205, USA.,Department of Neurology, University of Rochester Medical Center, Rochester, New York, USA
| |
Collapse
|
34
|
Genotype and phenotype analysis of 43 Iranian facioscapulohumeral muscular dystrophy patients; Evidence for anticipation. Neuromuscul Disord 2018; 28:303-314. [PMID: 29402602 DOI: 10.1016/j.nmd.2018.01.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 11/20/2017] [Accepted: 01/04/2018] [Indexed: 12/13/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is the third most common hereditary myopathy (prevalence 1/8300-1/20,000). It is typically characterized by progressive weakness of facial, scapular and humeral muscles. Pelvic, abdominal and lower limbs muscles may eventually be affected. FSHD is classified into two subgroups, FSHD1 and FSHD2. FSHD1 is due to a reduction in the copy number of D4Z4 macrosatellites on chromosome 4q35 (11-100 repeats in normal individuals and 1-10 repeats in patients), and FSHD2 is caused by mutations in SMCHD1 or DNMT3B. Here, we present clinical features and results of genetic analysis on 43 Iranian FSHD patients. Forty patients carried 2-7 D4Z4 repeats based on Southern blot analysis, thus confirming FSHD1 diagnosis in these patients. The number of patients with D4Z4 repeats in the range of 1-3, 4-6 and 7-9 were, respectively, 22, 17 and one. Patients with the lower number of D4Z4 repeats generally showed earlier onset and more severe disease presentations. Anticipation was observed in 14 multi-generational families. To the best of our knowledge, this is the first phenotype and genotype analysis of FSHD patients in the Iranian population. The results of this study will be beneficial for genetic counselling of FSHD patients and their families, and for the establishment of a simple affordable genetic test for Iranians as the majority of patients had 1-5 D4Z4 repeats.
Collapse
|
35
|
Potikanond S, Nimlamool W, Noordermeer J, Fradkin LG. Muscular Dystrophy Model. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1076:147-172. [PMID: 29951819 DOI: 10.1007/978-981-13-0529-0_9] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Muscular dystrophy (MD) is a group of muscle weakness disease involving in inherited genetic conditions. MD is caused by mutations or alteration in the genes responsible for the structure and functioning of muscles. There are many different types of MD which have a wide range from mild symptoms to severe disability. Some types involve the muscles used for breathing which eventually affect life expectancy. This chapter provides an overview of the MD types, its gene mutations, and the Drosophila MD models. Specifically, the Duchenne muscular dystrophy (DMD), the most common form of MD, will be thoroughly discussed including Dystrophin genes, their isoforms, possible mechanisms, and signaling pathways of pathogenesis.
Collapse
Affiliation(s)
- Saranyapin Potikanond
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Wutigri Nimlamool
- Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Jasprien Noordermeer
- Department of Molecular Biology, Leiden University Medical Center (LUMC), Leiden, The Netherlands
| | - Lee G Fradkin
- Department of Neurobiology, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
36
|
Lemmers RJ, van der Vliet PJ, Balog J, Goeman JJ, Arindrarto W, Krom YD, Straasheijm KR, Debipersad RD, Özel G, Sowden J, Snider L, Mul K, Sacconi S, van Engelen B, Tapscott SJ, Tawil R, van der Maarel SM. Deep characterization of a common D4Z4 variant identifies biallelic DUX4 expression as a modifier for disease penetrance in FSHD2. Eur J Hum Genet 2017; 26:94-106. [PMID: 29162933 DOI: 10.1038/s41431-017-0015-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 09/07/2017] [Accepted: 09/09/2017] [Indexed: 11/09/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy is caused by incomplete repression of the transcription factor DUX4 in skeletal muscle as a consequence of D4Z4 macrosatellite repeat contraction in chromosome 4q35 (FSHD1) or variants in genes encoding D4Z4 chromatin repressors (FSHD2). A clinical hallmark of FSHD is variability in onset and progression suggesting the presence of disease modifiers. A well-known cis modifier is the polymorphic DUX4 polyadenylation signal (PAS) that defines FSHD permissive alleles: D4Z4 chromatin relaxation on non-permissive alleles which lack the DUX4-PAS cannot cause disease in the absence of stable DUX4 mRNA. We have explored the nature and relevance of a common variant of the major FSHD haplotype 4A161, which is defined by 1.6 kb size difference of the most distal D4Z4 repeat unit. While the short variant (4A161S) has been extensively studied, we demonstrate that the long variant (4A161L) is relatively common in the European population, is capable of expressing DUX4, but that DUX4 mRNA processing differs from 4A161S. While we do not find evidence for a difference in disease severity between FSHD carriers of an 4A161S or 4A161L allele, our study does uncover biallelic DUX4 expression in FSHD2 patients. Compared to control individuals, we observed an increased frequency of FSHD2 patients homozygous for disease permissive alleles, and who are thus capable of biallelic DUX4 expression, while SMCHD1 variant carriers with only one permissive allele were significantly more often asymptomatic. This suggests that biallelic DUX4 expression lowers the threshold for disease presentation and is a modifier for disease severity in FSHD2.
Collapse
Affiliation(s)
- Richard Jlf Lemmers
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands.
| | | | - Judit Balog
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Jelle J Goeman
- Medical Statistics and Bioinformatics, Leiden University Medical Center, Leiden, Netherlands
| | - Wibowo Arindrarto
- Department of Molecular Epidemiology, Leiden University Medical Center, Leiden, Netherlands
| | - Yvonne D Krom
- Department of Neurology, Leiden University Medical Center, Leiden, Netherlands
| | | | - Rashmie D Debipersad
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Gizem Özel
- Department of Human Genetics, Leiden University Medical Center, Leiden, Netherlands
| | - Janet Sowden
- Neuromuscular Disease Unit, Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | - Lauren Snider
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Karlien Mul
- Neuromuscular Centre Nijmegen, Department of Neurology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Sabrina Sacconi
- Centre de référence des Maladies neuromusculaires and CNRS UMR6543, Nice University Hospital, Nice, France
| | - Baziel van Engelen
- Neuromuscular Centre Nijmegen, Department of Neurology, Radboud University Nijmegen Medical Centre, Nijmegen, Netherlands
| | - Stephen J Tapscott
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Rabi Tawil
- Neuromuscular Disease Unit, Department of Neurology, University of Rochester Medical Center, Rochester, NY, USA
| | | |
Collapse
|
37
|
DeSimone AM, Pakula A, Lek A, Emerson CP. Facioscapulohumeral Muscular Dystrophy. Compr Physiol 2017; 7:1229-1279. [PMID: 28915324 DOI: 10.1002/cphy.c160039] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Facioscapulohumeral Muscular Dystrophy is a common form of muscular dystrophy that presents clinically with progressive weakness of the facial, scapular, and humeral muscles, with later involvement of the trunk and lower extremities. While typically inherited as autosomal dominant, facioscapulohumeral muscular dystrophy (FSHD) has a complex genetic and epigenetic etiology that has only recently been well described. The most prevalent form of the disease, FSHD1, is associated with the contraction of the D4Z4 microsatellite repeat array located on a permissive 4qA chromosome. D4Z4 contraction allows epigenetic derepression of the array, and possibly the surrounding 4q35 region, allowing misexpression of the toxic DUX4 transcription factor encoded within the terminal D4Z4 repeat in skeletal muscles. The less common form of the disease, FSHD2, results from haploinsufficiency of the SMCHD1 gene in individuals carrying a permissive 4qA allele, also leading to the derepression of DUX4, further supporting a central role for DUX4. How DUX4 misexpression contributes to FSHD muscle pathology is a major focus of current investigation. Misexpression of other genes at the 4q35 locus, including FRG1 and FAT1, and unlinked genes, such as SMCHD1, has also been implicated as disease modifiers, leading to several competing disease models. In this review, we describe recent advances in understanding the pathophysiology of FSHD, including the application of MRI as a research and diagnostic tool, the genetic and epigenetic disruptions associated with the disease, and the molecular basis of FSHD. We discuss how these advances are leading to the emergence of new approaches to enable development of FSHD therapeutics. © 2017 American Physiological Society. Compr Physiol 7:1229-1279, 2017.
Collapse
Affiliation(s)
- Alec M DeSimone
- Wellstone Muscular Dystrophy Program, Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| | - Anna Pakula
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics and Genetics at Harvard Medical School, Boston, Massachusetts, USA
| | - Angela Lek
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, Massachusetts, USA.,Department of Pediatrics and Genetics at Harvard Medical School, Boston, Massachusetts, USA.,Australian Regenerative Medicine Institute, Monash University, Clayton, Victoria, Australia
| | - Charles P Emerson
- Wellstone Muscular Dystrophy Program, Department of Neurology, University of Massachusetts Medical School, Worcester, Massachusetts, USA
| |
Collapse
|
38
|
Long-term follow-up of MRI changes in thigh muscles of patients with Facioscapulohumeral dystrophy: A quantitative study. PLoS One 2017; 12:e0183825. [PMID: 28841698 PMCID: PMC5571945 DOI: 10.1371/journal.pone.0183825] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Accepted: 08/11/2017] [Indexed: 11/24/2022] Open
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is one of the most common hereditary muscular disorders. Currently FSHD has no known effective treatment and detailed data on the natural history are lacking. Determination of the efficacy of a given therapeutic approach might be difficult in FSHD given the slow and highly variable disease progression. Magnetic resonance imaging (MRI) has been widely used to qualitatively and quantitatively evaluate in vivo the muscle alterations in various neuromuscular disorders. The main aim of the present study was to investigate longitudinally the time-dependent changes occurring in thigh muscles of FSHD patients using quantitative MRI and to assess the potential relationships with the clinical findings. Thirty-five FSHD1 patients (17 females) were enrolled. Clinical assessment tools including manual muscle testing using medical research council score (MRC), and motor function measure (MFM) were recorded each year for a period ranging from 1 to 2 years. For the MRI measurements, we used a new quantitative index, i.e., the mean pixel intensity (MPI) calculated from the pixel-intensity distribution in T1 weighted images. The corresponding MPI scores were calculated for each thigh, for each compartment and for both thighs totally (MPItotal). The total mean pixel intensity (MPItotal) refers to the sum of each pixel signal intensity divided by the corresponding number of pixels. An increased MPItotal indicates both a raised fat infiltration together with a reduced muscle volume thereby illustrating disease progression. Clinical scores did not change significantly over time whereas MPItotal increased significantly from an initial averaged value of 39.6 to 41.1 with a corresponding rate of 0.62/year. While clinical scores and MPItotal measured at the start of the study were significantly related, no correlation was found between the rate of MPItotal and MRC sum score changes, MFMtotal and MFM subscores. The relative rate of MPItotal change was 2.3% (0.5–4.3)/year and was significantly higher than the corresponding rates measured for MRCS 0% (0–1.7) /year and MFMtotal 0% (0–2.0) /year (p = 0.000). On the basis of these results, we suggested that muscle MRI and more particularly the MPItotal index could be used as a reliable biomarker and outcome measure of disease progression. In slowly progressive myopathies such as FSHD, the MPItotal index might reveal subclinical changes, which could not be evidenced using clinical scales over a short period of time.
Collapse
|
39
|
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: 14] [Impact Index Per Article: 2.0] [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
|
40
|
Chagarlamudi H, Corbett A, Stoll M, Bibat G, Grosmann C, Matichak Stock C, Stinson N, Shapiro J, Wagner KR. Bone health in facioscapulohumeral muscular dystrophy: A cross-sectional study. Muscle Nerve 2017; 56:1108-1113. [DOI: 10.1002/mus.25619] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2017] [Indexed: 11/07/2022]
Affiliation(s)
- Hema Chagarlamudi
- Center for Genetic Muscle Disorders; Kennedy Krieger Institute; Baltimore Maryland USA
| | | | - Marion Stoll
- Molecular Medicine Laboratory; Concord Hospital; Concord New South Wales Australia
| | - Genila Bibat
- Center for Genetic Muscle Disorders; Kennedy Krieger Institute; Baltimore Maryland USA
| | - Carla Grosmann
- Center for Genetic Muscle Disorders; Kennedy Krieger Institute; Baltimore Maryland USA
- Rady Children's Hospital and Department of Neurosciences; University of California San Diego School of Medicine; San Diego California USA
| | - Carly Matichak Stock
- Center for Genetic Muscle Disorders; Kennedy Krieger Institute; Baltimore Maryland USA
| | - Nikia Stinson
- Center for Genetic Muscle Disorders; Kennedy Krieger Institute; Baltimore Maryland USA
| | - Jay Shapiro
- Osteogenesis Imperfecta Department; Kennedy Krieger Institute; Baltimore Maryland USA
| | - Kathryn R. Wagner
- Center for Genetic Muscle Disorders; Kennedy Krieger Institute; Baltimore Maryland USA
- Department of Neurology; The Johns Hopkins School of Medicine; Baltimore Maryland USA
- Department of Neuroscience; The Johns Hopkins School of Medicine; Baltimore Maryland USA
| |
Collapse
|
41
|
Sardone V, Zhou H, Muntoni F, Ferlini A, Falzarano MS. Antisense Oligonucleotide-Based Therapy for Neuromuscular Disease. Molecules 2017; 22:molecules22040563. [PMID: 28379182 PMCID: PMC6154734 DOI: 10.3390/molecules22040563] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/28/2017] [Accepted: 03/14/2017] [Indexed: 02/06/2023] Open
Abstract
Neuromuscular disorders such as Duchenne Muscular Dystrophy and Spinal Muscular Atrophy are neurodegenerative genetic diseases characterized primarily by muscle weakness and wasting. Until recently there were no effective therapies for these conditions, but antisense oligonucleotides, a new class of synthetic single stranded molecules of nucleic acids, have demonstrated promising experimental results and are at different stages of regulatory approval. The antisense oligonucleotides can modulate the protein expression via targeting hnRNAs or mRNAs and inducing interference with splicing, mRNA degradation, or arrest of translation, finally, resulting in rescue or reduction of the target protein expression. Different classes of antisense oligonucleotides are being tested in several clinical trials, and limitations of their clinical efficacy and toxicity have been reported for some of these compounds, while more encouraging results have supported the development of others. New generation antisense oligonucleotides are also being tested in preclinical models together with specific delivery systems that could allow some of the limitations of current antisense oligonucleotides to be overcome, to improve the cell penetration, to achieve more robust target engagement, and hopefully also be associated with acceptable toxicity. This review article describes the chemical properties and molecular mechanisms of action of the antisense oligonucleotides and the therapeutic implications these compounds have in neuromuscular diseases. Current strategies and carrier systems available for the oligonucleotides delivery will be also described to provide an overview on the past, present and future of these appealing molecules.
Collapse
Affiliation(s)
- Valentina Sardone
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK.
| | - Haiyan Zhou
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK.
| | - Francesco Muntoni
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK.
- MRC Centre for Neuromuscular Diseases, UCL Institute of Neurology, London WC1N 3BG, UK.
| | - Alessandra Ferlini
- Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neuroscience Programme, UCL Great Ormond Street Institute of Child Health, London WC1N 1EH, UK.
- UOL Medical Genetics, University of Ferrara, Ferrara 44121, Italy.
| | | |
Collapse
|
42
|
Ansseau E, Vanderplanck C, Wauters A, Harper SQ, Coppée F, Belayew A. Antisense Oligonucleotides Used to Target the DUX4 mRNA as Therapeutic Approaches in FaciosScapuloHumeral Muscular Dystrophy (FSHD). Genes (Basel) 2017; 8:genes8030093. [PMID: 28273791 PMCID: PMC5368697 DOI: 10.3390/genes8030093] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2016] [Revised: 02/16/2017] [Accepted: 02/22/2017] [Indexed: 02/02/2023] Open
Abstract
FacioScapuloHumeral muscular Dystrophy (FSHD) is one of the most prevalent hereditary myopathies and is generally characterized by progressive muscle atrophy affecting the face, scapular fixators; upper arms and distal lower legs. The FSHD locus maps to a macrosatellite D4Z4 repeat array on chromosome 4q35. Each D4Z4 unit contains a DUX4 gene; the most distal of which is flanked by a polyadenylation site on FSHD-permissive alleles, which allows for production of stable DUX4 mRNAs. In addition, an open chromatin structure is required for DUX4 gene transcription. FSHD thus results from a gain of function of the toxic DUX4 protein that normally is only expressed in germ line and stem cells. Therapeutic strategies are emerging that aim to decrease DUX4 expression or toxicity in FSHD muscle cells. We review here the heterogeneity of DUX4 mRNAs observed in muscle and stem cells; and the use of antisense oligonucleotides (AOs) targeting the DUX4 mRNA to interfere either with transcript cleavage/polyadenylation or intron splicing. We show in primary cultures that DUX4-targeted AOs suppress the atrophic FSHD myotube phenotype; but do not improve the disorganized FSHD myotube phenotype which could be caused by DUX4c over-expression. Thus; DUX4c might constitute another therapeutic target in FSHD.
Collapse
Affiliation(s)
- Eugénie Ansseau
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars 6, 7000-Mons, Belgium.
| | - Céline Vanderplanck
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars 6, 7000-Mons, Belgium.
| | - Armelle Wauters
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars 6, 7000-Mons, Belgium.
| | - Scott Q Harper
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH 43205, USA.
- Center for Gene Therapy, The Research Institute at Nationwide Children's Hospital, Columbus, OH 43205, USA.
| | - Frédérique Coppée
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars 6, 7000-Mons, Belgium.
| | - Alexandra Belayew
- Laboratory of Molecular Biology, Research Institute for Health Sciences and Technology, University of Mons, Avenue du Champ de Mars 6, 7000-Mons, Belgium.
| |
Collapse
|
43
|
Abstract
PURPOSE OF REVIEW This article describes the clinical characteristics, diagnosis, molecular pathogenesis, and treatment of facioscapulohumeral muscular dystrophy (FSHD). RECENT FINDINGS FSHD comprises two genetically distinct types that converge on a common downstream pathway of the expression of the toxic protein DUX4. Approximately 95% of patients have FSHD type 1 (FSHD1), in which loss of DNA repetitive elements (D4Z4 repeats) in the subtelomeric region of chromosome 4q causes decreased methylation and epigenetic derepression of DUX4, a gene contained within each D4Z4 repeat. FSHD type 2 (FSHD2) occurs through a deletion-independent mechanism but, similar to FSHD1, leads to decreased methylation and epigenetic derepression in the same region of chromosome 4q. Whereas FSHD1 is dominantly inherited, FSHD2 shows digenic inheritance, and about 80% of patients will have a mutation in the SMCHD1 gene. DUX4 lacks a polyadenylation signal, so both FSHD1 and FSHD2 only occur in the presence of permissive 4q polymorphisms, which provide a stabilizing polyadenylation sequence. FSHD is an epigenetic disease, and penetrance and severity are related to both the number of residual D4Z4 units and D4Z4 methylation. SUMMARY Recent consensus guidelines outline standards for care for FSHD, and identification of potential therapeutic targets have shifted emphasis in the research community toward drug development and clinical trial planning.
Collapse
|
44
|
Medication adherence in patients with myotonic dystrophy and facioscapulohumeral muscular dystrophy. J Neurol 2016; 263:2528-2537. [PMID: 27734165 DOI: 10.1007/s00415-016-8300-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 10/03/2016] [Accepted: 10/04/2016] [Indexed: 10/20/2022]
Abstract
Myotonic dystrophy (DM) and facioscapulohumeral muscular dystrophy (FSHD) are the two most common adult muscular dystrophies and have progressive and often disabling manifestations. Higher levels of medication adherence lead to better health outcomes, especially important to patients with DM and FSHD because of their multisystem manifestations and complexity of care. However, medication adherence has not previously been studied in a large cohort of DM type 1 (DM1), DM type 2 (DM2), and FSHD patients. The purpose of our study was to survey medication adherence and disease manifestations in patients enrolled in the NIH-supported National DM and FSHD Registry. The study was completed by 110 DM1, 49 DM2, and 193 FSHD patients. Notable comorbidities were hypertension in FSHD (44 %) and DM2 (37 %), gastroesophageal reflux disease in DM1 (24 %) and DM2 (31 %) and arrhythmias (29 %) and thyroid disease (20 %) in DM1. Each group reported high levels of adherence based on regimen complexity, medication costs, health literacy, side effect profile, and their beliefs about treatment. Only dysphagia in DM1 was reported to significantly impact medication adherence. Approximately 35 % of study patients reported polypharmacy (taking 6 or more medications). Of the patients with polypharmacy, the DM1 cohort was significantly younger (mean 55.0 years) compared to DM2 (59.0 years) and FSHD (63.2 years), and had shorter disease duration (mean 26 years) compared to FSHD (26.8 years) and DM2 (34.8 years). Future research is needed to assess techniques to ease pill swallowing in DM1 and to monitor polypharmacy and potential drug interactions in DM and FSHD.
Collapse
|
45
|
Fatehi F, Salort-Campana E, Le Troter A, Bendahan D, Attarian S. Muscle MRI of facioscapulohumeral dystrophy (FSHD): A growing demand and a promising approach. Rev Neurol (Paris) 2016; 172:566-571. [PMID: 27663058 DOI: 10.1016/j.neurol.2016.08.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 07/28/2016] [Accepted: 08/26/2016] [Indexed: 01/10/2023]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD), an inherited and progressive muscle disorder, is among the most common hereditary muscle disorders. From a clinical vantage point, FSHD is characterized by weakness of the facial, shoulder (often with scapular winging), arm (including biceps and triceps) and abdominal muscles. Forearm muscles are usually spared and weakness is usually asymmetrical. Over the past few decades, muscle magnetic resonance imaging (MRI) has become established as a reliable and accurate noninvasive tool for the diagnosis and assessment of progression in neuromuscular diseases, showing specific patterns of muscle involvement for a number of myopathies. More recently, MRI has been used to noninvasively identify quantitative biomarkers, allowing evaluation of the natural progression of disease and assessment of therapeutic interventions. In the present review, the intention was to present the most significant MRI developments related to diagnosis and pattern recognition in FSHD and to discuss its capacity to provide outcome measures.
Collapse
Affiliation(s)
- F Fatehi
- Reference center for Neuromuscular disorders and ALS, Timone University Hospital, Aix-Marseille University, 264, rue Saint-Pierre, 13385 Marseille cedex 05, France; Iranian Center of Neurological research and Shariati hospital, Neurology Department, Tehran University of Medical Sciences, Tehran, Iran
| | - E Salort-Campana
- Reference center for Neuromuscular disorders and ALS, Timone University Hospital, Aix-Marseille University, 264, rue Saint-Pierre, 13385 Marseille cedex 05, France; Aix-Marseille université, Inserm UMR S 910 Medical Genetics and Functional Genomics, 13385 Marseille, France
| | - A Le Troter
- Aix-Marseille université, centre de résonance magnétique biologique et médicale, UMR CNRS 7339, 13385 Marseille, France
| | - D Bendahan
- Aix-Marseille université, centre de résonance magnétique biologique et médicale, UMR CNRS 7339, 13385 Marseille, France
| | - S Attarian
- Reference center for Neuromuscular disorders and ALS, Timone University Hospital, Aix-Marseille University, 264, rue Saint-Pierre, 13385 Marseille cedex 05, France; Aix-Marseille université, Inserm UMR S 910 Medical Genetics and Functional Genomics, 13385 Marseille, France.
| |
Collapse
|
46
|
Trucco F, Pedemonte M, Fiorillo C, Tacchetti P, Brisca G, Bruno C, Minetti C. Respiratory pattern in a FSHD pediatric population. Respir Med 2016; 119:78-80. [PMID: 27692152 DOI: 10.1016/j.rmed.2016.08.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Revised: 06/07/2016] [Accepted: 08/21/2016] [Indexed: 10/21/2022]
Abstract
Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant inherited disorder characterized by selective weakness of face and upper arms and girdle. Respiratory involvement in FSHD has been described mainly in the most severely affected patients. In this work we tested the respiratory function by spirometry in 12 patients affected by FSHD with onset before 18 years. Spirometry results were correlated with motor involvement and compared to aged matched group of Becker patients. Of note FSHD patients present a peculiar pattern characterized by a flat shape in flow-volume loop. Respiratory volumes correlate with clinical severity and expiratory phase is specifically affected in comparison to other muscular dystrophies.
Collapse
Affiliation(s)
- Federica Trucco
- Pediatric Neurology and Muscle Disease Unit, Istituto Giannina Gaslini, Genova, Italy.
| | - Marina Pedemonte
- Pediatric Neurology and Muscle Disease Unit, Istituto Giannina Gaslini, Genova, Italy
| | - Chiara Fiorillo
- Molecular Medicine, IRCCS Stella Maris, Via dei Giacinti 2, 56128, Pisa, Italy
| | - Paola Tacchetti
- Pediatric Neurology and Muscle Disease Unit, Istituto Giannina Gaslini, Genova, Italy
| | - Giacomo Brisca
- Pediatric Neurology and Muscle Disease Unit, Istituto Giannina Gaslini, Genova, Italy
| | - Claudio Bruno
- Center of Myology and Neurodegenerative Disorders, Department of Neuroscience, Istituto Giannina Gaslini, Genova, Italy; Pediatric Neurology and Muscle Disease Unit, Istituto Giannina Gaslini, Genova, Italy
| | - Carlo Minetti
- Pediatric Neurology and Muscle Disease Unit, Istituto Giannina Gaslini, Genova, Italy; Department of Pediatrics, University of Genova, Italy
| |
Collapse
|
47
|
Bakker M, Schipper K, Geurts AC, Abma TA. It's not just physical: a qualitative study regarding the illness experiences of people with facioscapulohumeral muscular dystrophy. Disabil Rehabil 2016; 39:978-986. [PMID: 27211201 DOI: 10.3109/09638288.2016.1172673] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
PURPOSE Little is known about the illness experiences of people with Facioscapulohumeral Muscular Dystrophy (FSHD). The aim of this study was to provide insight into the illness experiences of people with FSHD in order to tailor rehabilitation programs to individual needs and expectations. METHODS Twenty-five semi-structured interviews were conducted with people with FSHD. The interviews were audiotaped, transcribed and member checked. Computerized (MAXqda) and manual techniques were used for thematic data analysis. RESULTS Intra- as well as extra-individual aspects play a role in the illness experiences of people with FSHD. Integrating the consequences of the diagnosis and symptoms, coping with heredity and progenity, adjusting to a decreasing independence, and the accompanying changing relationship with one's partner, are mentioned as intra-individual aspects. As extra-individual factors are the responses of the social environment, which was mentioned as well as used assistive devices, and maintaining or giving up work. CONCLUSIONS Better understanding of the individual illness experiences, cognitions, and social context of people with FSHD can give health professionals tools to improve their care and give researchers direction for future studies to evaluate healthcare improvements from a holistic, patient-centred perspective. Implications for Rehabilitation FSHD has a major impact on people's lives. Besides the physical consequences, issues such as heredity, progenity, changing (intimate) relationships, social interactions and work should be addressed by rehabilitation professionals. Dependent on the timing of the diagnosis (early or later in life) people with FSHD could, in addition to medical consultation and physical therapy, profit from support by a social worker, occupational therapist and/or genetic Counselor for the above-mentioned themes to be addressed more extensively. It is relevant for rehabilitation professionals to become familiar with the personal characteristics and social circumstances of the patient before communicating the diagnosis and prognosis in order to individually tailor the content of the communication.
Collapse
Affiliation(s)
- Minne Bakker
- a Department of Medical Humanities , VU University Medical Centre , Amsterdam , The Netherlands
| | - Karen Schipper
- a Department of Medical Humanities , VU University Medical Centre , Amsterdam , The Netherlands
| | - Alexander C Geurts
- b Department of Rehabilitation , Radboud University Medical Centre, Donders Centre for Neuroscience , Amsterdam , The Netherlands
| | - Tineke A Abma
- a Department of Medical Humanities , VU University Medical Centre , Amsterdam , The Netherlands
| |
Collapse
|
48
|
Design, set-up and utility of the UK facioscapulohumeral muscular dystrophy patient registry. J Neurol 2016; 263:1401-8. [PMID: 27159994 PMCID: PMC4929161 DOI: 10.1007/s00415-016-8132-1] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 04/12/2016] [Accepted: 04/12/2016] [Indexed: 12/21/2022]
Abstract
Facioscapulohumeral dystrophy (FSHD) is a rare inherited neuromuscular disease estimated to affect 1/15,000 people. Through basic research, remarkable progress has been made towards the development of targeted therapies. Patient identification, through registries or other means is essential for trial-readiness. The UK FSHD Patient Registry is a patient initiated registry that collects standardised and internationally agreed dataset of self-reported clinical details combined with professionally verified genetic information. It includes four additional questionnaires to capture patient reported outcomes related to pain, quality of life and scapular fixation. Between 2013 and 2015, 518 patients registered 243 males, 241 females with a mean age of 47.8 years. Most of the patients have FSHD type 1 (91.7 %), and weakness of the facial (59.2 %) was the most prevalent symptom at onset, followed by shoulder-girdle muscles (53.3 %) and distal (22.45 %) or proximal lower limb weakness (14.8 %). 85.57 % patients were ambulant or ambulant with assistance at the time of registration, 7.9 % report respiratory insufficiency. The registry has demonstrated utility with the recruitment of patients for a natural history study of infantile onset FSHD, and the longitudinal analysis of patient-related outcomes will provide much-needed baseline information to power future trials. The internationally agreed core dataset enables national registries to participate in a “Global FSHD registry”. We suggest that the registry’s ability to interoperate with other large datasets will be instrumental for sharing and exploiting data globally.
Collapse
|
49
|
|