Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 80
Filtrar
1.
J Neuromuscul Dis ; 11(2): 459-472, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38277300

RESUMO

Background: Facioscapulohumeral muscular dystrophy (FSHD) is a genetic disorder characterized by progressive muscle weakness leading to permanent disability. There are no curative treatments, however, there are several upcoming clinical trials testing new therapies in FSHD. Objective: This study aimed to explore the disease burden and patient preferences of people with FSHD to ensure that clinical trials can be designed to include outcome measures that are relevant and important to patients. Methods: A survey was developed with a steering committee clinicians and physiotherapists with relevant experience in the disease, patient representatives, a registry expert and industry consultants. Themes of the survey included; participant demographics, disease progression and impact on function, factors encouraging or discouraging clinical trial participation, and positive outcomes of a clinical trial. Results: 1147 participants responded to the online survey, representing 26 countries across Europe and a range of disease severities. The study highlighted the key symptoms causing concern for FSHD patients - muscle weakness and mobility issues - reflecting what participants want targeted for future therapies. The need for clear information and communication throughout clinical trials was emphasised. Factors most encouraging trial participation included access to new investigational therapies, access to trial results and benefits for the FSHD community. Factors most discouraging trial participation included travel related issues and fear of side effects. Conclusions: The results from this study identify the patient reported burden of FSHD and should provide researchers and industry with areas of therapeutic research that would be meaningful to patients, as well as supporting the development of patient centric outcome measures in clinical trials.


Assuntos
Distrofia Muscular Facioescapuloumeral , Humanos , Distrofia Muscular Facioescapuloumeral/terapia , Viagem , Doença Relacionada a Viagens , Efeitos Psicossociais da Doença , Debilidade Muscular , Medidas de Resultados Relatados pelo Paciente
2.
Neuromuscul Disord ; 35: 53-57, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-37978033

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is one of the most common inherited muscular dystrophies. As part of the FSHD Society's commitment to promote global communication and collaboration among researchers, the Society collaborated with FSHD Europe and convened its 30th annual International Research Congress (IRC) on June 15-16, 2023, in the city of Milan, Italy. Over 240 researchers, clinicians, patients and pharmaceutical company representatives from a wide geographical background participated to hear about the latest developments and breakthroughs in the field. The meeting was structured to provide a mix of basic and clinical research in five sessions: 1. Discovery research & genetics; 2. Outcome assessments; 3. Disease mechanisms & interventional strategies; 4. Clinical studies & trial design; and 5. Pediatric FSHD. The keynote speakers were Professor Baziel van Engelen (on the importance of incorporating the patient's voice to help refine and improve basic laboratory and clinical research) and Dr. Bénédict Chazaud (on the role of the immune system in normal muscle regeneration and in Duchenne muscular dystrophy). The FSHD IRC was preceded by the Industry Collaborative for Therapeutic Development in FSHD meeting and followed by the World FSHD Alliance network of national patient groups and advocacy organizations for FSHD summit. The Congress concluded with the announcement for the 2024 International Research Congress, which will take place on June 13-14, 2024 in Denver, Colorado, USA, and followed by the FSHD Society's flagship educational conference for the FSHD community, the Patient Connect Conference, on June 15-16, 2024.


Assuntos
Distrofia Muscular de Duchenne , Distrofia Muscular Facioescapuloumeral , Humanos , Criança , Distrofia Muscular Facioescapuloumeral/terapia , Itália , Europa (Continente)
3.
Muscle Nerve ; 68(4): 422-431, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37610084

RESUMO

INTRODUCTION/AIMS: As promising therapeutic interventions are tested among patients with facioscapulohumeral muscular dystrophy (FSHD), there is a clear need for valid and reliable outcome tools to track disease progression and therapeutic gain in clinical trials and for clinical monitoring. Our aim was to develop and validate the Facioscapulohumeral Muscular Dystrophy-Health Index (FSHD-HI) as a multifaceted patient-reported outcome measure (PRO) designed to measure disease burden in adults with FSHD. METHODS: Through initial interviews with 20 individuals and a national cross-sectional study with 328 individuals with FSHD, we identified the most prevalent and impactful symptoms in FSHD. The most relevant symptoms were included in the FSHD-HI. We used patient interviews, test-retest reliability evaluation, known groups validity testing, and factor analysis to evaluate and optimize the FSHD-HI. RESULTS: The FSHD-HI contains 14 subscales that measure FSHD disease burden from the patient's perspective. Fourteen adults with FSHD participated in semistructured beta interviews and found the FSHD-HI to be clear, usable, and relevant to them. Thirty-two adults with FSHD participated in test-retest reliability assessments, which demonstrated the high reliability of the FSHD-HI total score (intraclass correlation coefficient = 0.924). The final FSHD-HI and its subscales also demonstrated a high internal consistency (Cronbach α = 0.988). DISCUSSION: The FSHD-HI provides researchers and clinicians with a reliable and valid mechanism to measure multifaceted disease burden in patients with FSHD. The FSHD-HI may facilitate quantification of therapeutic effectiveness, as demonstrated by its use as a secondary and exploratory measure in several clinical trials.


Assuntos
Distrofia Muscular Facioescapuloumeral , Adulto , Humanos , Estudos Transversais , Distrofia Muscular Facioescapuloumeral/diagnóstico , Distrofia Muscular Facioescapuloumeral/terapia , Reprodutibilidade dos Testes , Efeitos Psicossociais da Doença , Progressão da Doença
4.
Int J Mol Sci ; 24(11)2023 May 30.
Artigo em Inglês | MEDLINE | ID: mdl-37298453

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) represents the third most common form of muscular dystrophy and is characterized by muscle weakness and atrophy. FSHD is caused by the altered expression of the transcription factor double homeobox 4 (DUX4), which is involved in several significantly altered pathways required for myogenesis and muscle regeneration. While DUX4 is normally silenced in the majority of somatic tissues in healthy individuals, its epigenetic de-repression has been linked to FSHD, resulting in DUX4 aberrant expression and cytotoxicity in skeletal muscle cells. Understanding how DUX4 is regulated and functions could provide useful information not only to further understand FSHD pathogenesis, but also to develop therapeutic approaches for this disorder. Therefore, this review discusses the role of DUX4 in FSHD by examining the possible molecular mechanisms underlying the disease as well as novel pharmacological strategies targeting DUX4 aberrant expression.


Assuntos
Distrofia Muscular Facioescapuloumeral , Humanos , Regulação da Expressão Gênica , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/terapia , Distrofia Muscular Facioescapuloumeral/metabolismo
6.
Nat Rev Neurol ; 19(2): 91-108, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36627512

RESUMO

Advances in the molecular understanding of facioscapulohumeral muscular dystrophy (FSHD) have revealed that FSHD results from epigenetic de-repression of the DUX4 gene in skeletal muscle, which encodes a transcription factor that is active in early embryonic development but is normally silenced in almost all somatic tissues. These advances also led to the identification of targets for disease-altering therapies for FSHD, as well as an improved understanding of the molecular mechanism of the disease and factors that influence its progression. Together, these developments led the FSHD research community to shift its focus towards the development of disease-modifying treatments for FSHD. This Review presents advances in the molecular and clinical understanding of FSHD, discusses the potential targeted therapies that are currently being explored, some of which are already in clinical trials, and describes progress in the development of FSHD-specific outcome measures and assessment tools for use in future clinical trials.


Assuntos
Distrofia Muscular Facioescapuloumeral , Humanos , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/terapia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Músculo Esquelético/metabolismo , Regulação da Expressão Gênica
8.
Methods Mol Biol ; 2587: 197-208, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36401032

RESUMO

The third most common muscular dystrophy in the world, facioscapulohumeral muscular dystrophy (FSHD), is an inherited disorder characterized by distinct asymmetric, progressive skeletal muscle weakness that begins in the upper body and spreads to other regions with age. It is caused by mutations that induce aberrant expression of the DUX4 gene in skeletal muscle. DUX4 is highly cytotoxic in skeletal muscle, dysregulating numerous signaling pathways as a result of its transcription factor activity. A promising set of approaches being developed to treat FSHD uses antisense oligonucleotides (AOs) to inhibit DUX4 transcript expression. Both steric-blocking and gapmer AOs have been shown to induce efficient DUX4 transcript knockdown in vitro and in vivo. Here, we describe a protocol that allows reliable screening of DUX4-targeting AOs through the evaluation of DUX4 transcript expression by quantitative real-time polymerase chain reaction. We also describe methods to assess the efficacy of these AOs by looking at their effect on the expression of DUX4 downstream target and potential off-target genes, as well as on the amelioration of in vitro muscle cell phenotypes.


Assuntos
Distrofia Muscular Facioescapuloumeral , Humanos , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/terapia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Genes Homeobox , Músculo Esquelético/metabolismo , Células Musculares/metabolismo
9.
Continuum (Minneap Minn) ; 28(6): 1735-1751, 2022 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-36537978

RESUMO

PURPOSE OF REVIEW: This article reviews the current knowledge on the clinical characteristics and disease mechanism of facioscapulohumeral muscular dystrophy (FSHD), as well as advances in targeted therapy development. RECENT FINDINGS: FSHD has a wide range of severity, yet a distinct phenotype characterized by weakness of the facial, shoulder, and upper arm muscles, followed by weakness of the trunk and leg muscles. It can be caused by two genetic mechanisms that share a common downstream pathway, namely, the epigenetic derepression and subsequent misexpression of the myotoxic DUX4 transcription factor. Treatment is currently supportive and outlined in evidence-based guidelines. Advances in the understanding of the pathogenic mechanism of FSHD are paving the way for targeted therapy development. Approaches for targeted therapies to reduce DUX4 expression that are currently being explored include small molecules, antisense oligonucleotides, vector-based RNA interference, and gene therapy. In anticipation of more clinical trials, "clinical trial preparedness," including the development of sensitive biomarkers and clinical outcome measures, are needed. SUMMARY: The cornerstones of the diagnosis of FSHD are clinical observation and genetic testing. Management is currently supportive, but progress in the understanding of the disease mechanism has shifted the field of FSHD toward targeted therapy development.


Assuntos
Distrofia Muscular Facioescapuloumeral , Humanos , Distrofia Muscular Facioescapuloumeral/diagnóstico , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/terapia , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Fenótipo , Biomarcadores/metabolismo
10.
Phys Occup Ther Pediatr ; 42(6): 680-689, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35473461

RESUMO

AIMS: This study aims to investigate the feasibility and safety of short-term functional electrical stimulation (FES) training of the quadriceps femoris muscles in a child with facioscapulohumeral muscular dystrophy (FSHD). METHODS: A 7-year-old child with FSHD received treatment due to a decrease in functional performance and difficulty climbing stairs. The child was followed up with a home-based exercise program. FES was applied twice a week during stair climbing for six weeks. Muscle activation of the quadriceps femoris was measured using superficial electromyography, muscle strength was measured with a hand-held dynamometer, and functional performance was assessed with the 6-Minute Walk and the Stair Climb Tests before and after the treatment period. RESULTS: At the end of the treatment, there was an improvement in muscle activation. While muscle strength increased in the quadriceps femoris muscle of the non-dominant side, it remained constant on the dominant side. Functional performance test results also improved. CONCLUSIONS: FES was a feasible and safe tool to use in our case, a child with FSHD.


Assuntos
Distrofia Muscular Facioescapuloumeral , Criança , Estimulação Elétrica , Terapia por Exercício , Estudos de Viabilidade , Humanos , Força Muscular/fisiologia , Distrofia Muscular Facioescapuloumeral/terapia
11.
Cells ; 11(4)2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35203336

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is a debilitating muscular dystrophy with a variable age of onset, severity, and progression. While there is still no cure for this disease, progress towards FSHD therapies has accelerated since the underlying mechanism of epigenetic derepression of the double homeobox 4 (DUX4) gene leading to skeletal muscle toxicity was identified. This has facilitated the rapid development of novel therapies to target DUX4 expression and downstream dysregulation that cause muscle degeneration. These discoveries and pre-clinical translational studies have opened new avenues for therapies that await evaluation in clinical trials. As the field anticipates more FSHD trials, the need has grown for more reliable and quantifiable outcome measures of muscle function, both for early phase and phase II and III trials. Advanced tools that facilitate longitudinal clinical assessment will greatly improve the potential of trials to identify therapeutics that successfully ameliorate disease progression or permit muscle functional recovery. Here, we discuss current and emerging FSHD outcome measures and the challenges that investigators may experience in applying such measures to FSHD clinical trial design and implementation.


Assuntos
Distrofia Muscular Facioescapuloumeral , Proteínas de Homeodomínio/metabolismo , Humanos , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapuloumeral/tratamento farmacológico , Distrofia Muscular Facioescapuloumeral/terapia , Avaliação de Resultados em Cuidados de Saúde
12.
Hum Gene Ther ; 33(17-18): 923-935, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35078334

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is a rare muscle dystrophy causing muscle weakness initially in the face, shoulders, and upper arms, and extends 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. In this study, 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.


Assuntos
Distrofia Muscular Facioescapuloumeral , Animais , Diafragma/metabolismo , Diafragma/patologia , Modelos Animais de Doenças , Proteínas de Homeodomínio/genética , Camundongos , Camundongos Transgênicos , Músculo Esquelético/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/metabolismo , Distrofia Muscular Facioescapuloumeral/terapia
13.
Elife ; 112022 01 25.
Artigo em Inglês | MEDLINE | ID: mdl-35076017

RESUMO

Skeletal muscle myoblasts (iMyoblasts) were generated from human induced pluripotent stem cells (iPSCs) using an efficient and reliable transgene-free induction and stem cell selection protocol. Immunofluorescence, flow cytometry, qPCR, digital RNA expression profiling, and scRNA-Seq studies identify iMyoblasts as a PAX3+/MYOD1+ skeletal myogenic lineage with a fetal-like transcriptome signature, distinct from adult muscle biopsy myoblasts (bMyoblasts) and iPSC-induced muscle progenitors. iMyoblasts can be stably propagated for >12 passages or 30 population doublings while retaining their dual commitment for myotube differentiation and regeneration of reserve cells. iMyoblasts also efficiently xenoengrafted into irradiated and injured mouse muscle where they undergo differentiation and fetal-adult MYH isoform switching, demonstrating their regulatory plasticity for adult muscle maturation in response to signals in the host muscle. Xenograft muscle retains PAX3+ muscle progenitors and can regenerate human muscle in response to secondary injury. As models of disease, iMyoblasts from individuals with Facioscapulohumeral Muscular Dystrophy revealed a previously unknown epigenetic regulatory mechanism controlling developmental expression of the pathological DUX4 gene. iMyoblasts from Limb-Girdle Muscular Dystrophy R7 and R9 and Walker Warburg Syndrome patients modeled their molecular disease pathologies and were responsive to small molecule and gene editing therapeutics. These findings establish the utility of iMyoblasts for ex vivo and in vivo investigations of human myogenesis and disease pathogenesis and for the development of muscle stem cell therapeutics.


Muscular dystrophies are a group of inherited genetic diseases characterised by progressive muscle weakness. They lead to disability or even death, and no cure exists against these conditions. Advances in genome sequencing have identified many mutations that underly muscular dystrophies, opening the door to new therapies that could repair incorrect genes or rebuild damaged muscles. However, testing these ideas requires better ways to recreate human muscular dystrophy in the laboratory. One strategy for modelling muscular dystrophy involves coaxing skin or other cells from an individual into becoming 'induced pluripotent stem cells'; these can then mature to form almost any adult cell in the body, including muscles. However, this approach does not usually create myoblasts, the 'precursor' cells that specifically mature into muscle during development. This limits investigations into how disease-causing mutations impact muscle formation early on. As a response, Guo et al. developed a two-step protocol of muscle maturation followed by stem cell growth selection to isolate and grow 'induced myoblasts' from induced pluripotent stem cells taken from healthy volunteers and muscular dystrophy patients. These induced myoblasts can both make more of themselves and become muscle, allowing Guo et al. to model three different types of muscular dystrophy. These myoblasts also behave as stem cells when grafted inside adult mouse muscles: some formed human muscle tissue while others remained as precursor cells, which could then respond to muscle injury and start repair. The induced myoblasts developed by Guo et al. will enable scientists to investigate the impacts of different mutations on muscle tissue and to better test treatments. They could also be used as part of regenerative medicine therapies, to restore muscle cells in patients.


Assuntos
Células-Tronco Pluripotentes Induzidas/transplante , Distrofia Muscular Facioescapuloumeral/terapia , Mioblastos/transplante , Animais , Diferenciação Celular , Linhagem Celular , Linhagem da Célula , Células Cultivadas , Modelos Animais de Doenças , Proteínas de Homeodomínio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Camundongos , Desenvolvimento Muscular , Distrofia Muscular Facioescapuloumeral/patologia , Fator de Transcrição PAX3/metabolismo , Recuperação de Função Fisiológica , Regeneração
14.
J Neurol ; 268(12): 4778-4788, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34043041

RESUMO

BACKGROUND: Promising genetic therapies are being investigated in facioscapulohumeral muscular dystrophy (FSHD). However, the current cost of illness is largely unknown. OBJECTIVE: This study aimed at determining the socioeconomic burden of FSHD. METHODS: Adult patients with FSHD from the Dutch FSHD registry were invited to complete a questionnaire on medical consumption, work productivity and health-related quality of life (HR-QoL) using the EQ-5D-5L. Associated costs were calculated from a societal perspective. A generalized linear model was fitted to the data to investigate whether level of mobility was related to annual costs of illness. RESULTS: 172 patients with FSHD completed the questionnaire (response rate 65%). The per-patient annual direct medical costs of FSHD were estimated at €12,077, direct non-medical costs at €9179 and indirect costs at €5066, adding up to a total cost of illness of €26,322 per patient per year. The direct costs of illness were €21,256, approximately five times higher than the mean per-capita health expenditures in the Netherlands. Major cost-driving factors were formal home care and informal care. A decreased level of mobility was associated with higher direct costs of illness. HR-QoL was significantly reduced in patients with FSHD with a median health utility value of 0.63. CONCLUSIONS: We show that FSHD is associated with substantial direct and indirect socioeconomic costs as well as a reduction in HR-QoL. These findings are important for health care decision makers and aids in allocation of research funds and evaluation of the cost-effectiveness of novel therapies.


Assuntos
Distrofia Muscular Facioescapuloumeral , Qualidade de Vida , Adulto , Efeitos Psicossociais da Doença , Estudos Transversais , Humanos , Distrofia Muscular Facioescapuloumeral/epidemiologia , Distrofia Muscular Facioescapuloumeral/terapia , Fatores Socioeconômicos , Inquéritos e Questionários
15.
Trends Mol Med ; 27(6): 588-601, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33863674

RESUMO

DUX4, a gene encoding a transcription factor involved in early embryogenesis, is located within the D4Z4 subtelomeric repeat on chromosome 4q35. In most healthy somatic tissues, DUX4 is heavily repressed by multiple genetic and epigenetic mechanisms, and its aberrant expression is linked to facioscapulohumeral muscular dystrophy (FSHD) where it has been extensively studied. Recently, DUX4 expression has been implicated in oncogenesis, although this is much less explored. In this review, we discuss multiple levels of control of DUX4 expression, including enhancer-promoter interactions, DNA methylation, histone modifications, noncoding RNAs, and telomere positioning effect. We also connect disparate data on intrachromosomal contacts involving DUX4 and emphasize the feedback loops in DUX4 regulation. Finally, we bridge data on DUX4 in FSHD and cancer and discuss prospective approaches for future FSHD therapies and the potential outcomes of DUX4 inhibition in cancer.


Assuntos
Epigênese Genética , Regulação da Expressão Gênica , Proteínas de Homeodomínio/metabolismo , Terapia de Alvo Molecular , Distrofia Muscular Facioescapuloumeral/patologia , Neoplasias/patologia , Metilação de DNA , Inativação Gênica , Proteínas de Homeodomínio/genética , Humanos , Distrofia Muscular Facioescapuloumeral/metabolismo , Distrofia Muscular Facioescapuloumeral/terapia , Neoplasias/metabolismo , Neoplasias/terapia
16.
Trends Mol Med ; 27(2): 123-137, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33092966

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is one of the most common types of muscular dystrophy, affecting roughly one in 8000 individuals. The complex underlying genetics and poor mechanistic understanding has caused a bottleneck in therapeutic development. Until the discovery of DUX4 and its causal role in FSHD, most trials were untargeted with limited results. Emerging approaches can learn from these early trials to increase their chance of success. Here, we explore the evolution of FSHD clinical trials from nonspecific anabolic or anti-inflammatory/oxidant strategies to cutting-edge molecular therapies targeting DUX4, and we discuss the importance of clinical outcome measures. With combined advances across multiple facets of FSHD research, the field is now poised to accelerate the process of therapeutic discovery and testing.


Assuntos
Distrofia Muscular Facioescapuloumeral/terapia , Animais , Biomarcadores , Estudos Clínicos como Assunto , Terapia Combinada , Gerenciamento Clínico , Suscetibilidade a Doenças , Desenvolvimento de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Predisposição Genética para Doença , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/metabolismo , Humanos , Terapia de Alvo Molecular , Distrofia Muscular Facioescapuloumeral/diagnóstico , Distrofia Muscular Facioescapuloumeral/etiologia , Distrofia Muscular Facioescapuloumeral/metabolismo , Oxirredução , Resultado do Tratamento
17.
Mol Ther ; 29(2): 848-858, 2021 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-33068777

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD) is an autosomal dominant disorder characterized by a progressive, asymmetric weakening of muscles, starting with those in the upper body. It is caused by aberrant expression of the double homeobox protein 4 gene (DUX4) in skeletal muscle. FSHD is currently incurable. We propose to develop a therapy for FSHD using antisense 2'-O-methoxyethyl (2'-MOE) gapmers, to knock down DUX4 mRNA expression. Using immortalized patient-derived muscle cells and local intramuscular injections in the FLExDUX4 FSHD mouse model, we showed that our designed 2'-MOE gapmers significantly reduced DUX4 transcript levels in vitro and in vivo, respectively. Furthermore, in vitro, we observed significantly reduced expression of DUX4-activated downstream targets, restoration of FSHD signature genes by RNA sequencing, significant improvements in myotube morphology, and minimal off-target activity. This work facilitates the development of a promising candidate therapy for FSHD and lays down the foundation for in vivo systemic treatment studies.


Assuntos
Técnicas de Silenciamento de Genes , Inativação Gênica , Terapia Genética , Proteínas de Homeodomínio/genética , Distrofia Muscular Facioescapuloumeral/genética , Distrofia Muscular Facioescapuloumeral/terapia , Oligonucleotídeos Antissenso , Animais , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Músculo Esquelético/metabolismo
18.
Int J Mol Sci ; 21(20)2020 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-33096728

RESUMO

Facioscapulohumeral muscular dystrophy (FSHD)-the worldwide third most common inherited muscular dystrophy caused by the heterozygous contraction of a 3.3 kb tandem repeat (D4Z4) on a chromosome with a 4q35 haplotype-is a progressive genetic myopathy with variable onset of symptoms, distribution of muscle weakness, and clinical severity. While much is known about the clinical course of adult FSHD, data on the early-onset infantile phenotype, especially on the progression of the disease, are relatively scarce. Contrary to the classical form, patients with infantile FSHD more often have a rapid decline in muscle wasting and systemic features with multiple extramuscular involvements. A rough correlation between the phenotypic severity of FSHD and the D4Z4 repeat size has been reported, and the majority of patients with infantile FSHD obtain a very short D4Z4 repeat length (one to three copies, EcoRI size 10-14 kb), in contrast to the classical, slowly progressive, form of FSHD (15-38 kb). With the increasing identifications of case reports and the advance in genetic diagnostics, recent studies have suggested that the infantile variant of FSHD is not a genetically separate entity but a part of the FSHD spectrum. Nevertheless, many questions about the clinical phenotype and natural history of infantile FSHD remain unanswered, limiting evidence-based clinical management. In this review, we summarize the updated research to gain insight into the clinical spectrum of infantile FSHD and raise views to improve recognition and understanding of its underlying pathomechanism, and further, to advance novel treatments and standard care methods.


Assuntos
Distrofia Muscular Facioescapuloumeral/etiologia , Distrofia Muscular Facioescapuloumeral/terapia , Idade de Início , Humanos , Distrofia Muscular Facioescapuloumeral/patologia , Retina/fisiopatologia
19.
Int J Mol Sci ; 21(15)2020 Aug 04.
Artigo em Inglês | MEDLINE | ID: mdl-32759720

RESUMO

Deficits in plasma membrane repair have been identified in dysferlinopathy and Duchenne Muscular Dystrophy, and contribute to progressive myopathy. Although Facioscapulohumeral Muscular Dystrophy (FSHD) shares clinicopathological features with these muscular dystrophies, it is unknown if FSHD is characterized by plasma membrane repair deficits. Therefore, we exposed immortalized human FSHD myoblasts, immortalized myoblasts from unaffected siblings, and myofibers from a murine model of FSHD (FLExDUX4) to focal, pulsed laser ablation of the sarcolemma. Repair kinetics and success were determined from the accumulation of intracellular FM1-43 dye post-injury. We subsequently treated FSHD myoblasts with a DUX4-targeting antisense oligonucleotide (AON) to reduce DUX4 expression, and with the antioxidant Trolox to determine the role of DUX4 expression and oxidative stress in membrane repair. Compared to unaffected myoblasts, FSHD myoblasts demonstrate poor repair and a greater percentage of cells that failed to repair, which was mitigated by AON and Trolox treatments. Similar repair deficits were identified in FLExDUX4 myofibers. This is the first study to identify plasma membrane repair deficits in myoblasts from individuals with FSHD, and in myofibers from a murine model of FSHD. Our results suggest that DUX4 expression and oxidative stress may be important targets for future membrane-repair therapies.


Assuntos
Proteínas de Homeodomínio/genética , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular Facioescapuloumeral/genética , Estresse Oxidativo/genética , Adulto , Idoso , Animais , Antioxidantes/metabolismo , Membrana Celular/genética , Membrana Celular/metabolismo , Células Cultivadas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/antagonistas & inibidores , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Fibras Musculares Esqueléticas/patologia , Distrofia Muscular Facioescapuloumeral/metabolismo , Distrofia Muscular Facioescapuloumeral/patologia , Distrofia Muscular Facioescapuloumeral/terapia , Mioblastos/metabolismo , Miofibrilas/genética , Miofibrilas/metabolismo , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Estresse Oxidativo/efeitos dos fármacos
20.
Schmerz ; 34(5): 447-459, 2020 Oct.
Artigo em Alemão | MEDLINE | ID: mdl-32734404

RESUMO

In this article we address the relevance of rare diseases and their peculiarities with respect to pain therapy. Towards this end, four rare diseases (hemophilia, Morbus Fabry, dermatomyositis, and facioscapulohumeral dystrophy (FSHD)) will be presented and fundamental aspects of their pain therapies described. The diseases were chosen to showcase a pain therapy based on the WHO-step-by-step plan (hemophilia), a complex but established pain therapy (M. Fabry), and two less well established, individually adapted pain therapies (dermatomyositis, FSHD).


Assuntos
Distrofia Muscular Facioescapuloumeral , Manejo da Dor , Doenças Raras , Humanos , Distrofia Muscular Facioescapuloumeral/terapia , Doenças Raras/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...