Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 50
Filtrar
1.
Nat Med ; 30(1): 199-206, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38177855

RESUMO

Limb-girdle muscular dystrophy 2E/R4 is caused by mutations in the ß-sarcoglycan (SGCB) gene, leading to SGCB deficiency and consequent muscle loss. We developed a gene therapy approach based on functional replacement of the deficient SCB protein. Here we report interim results from a first-in-human, open-label, nonrandomized, phase 1/2 trial evaluating the safety and efficacy of bidridistrogene xeboparvovec, an adeno-associated virus-based gene therapy containing a codon-optimized, full-length human SGCB transgene. Patients aged 4-15 years with confirmed SGCB mutations at both alleles received one intravenous infusion of either 1.85 × 1013 vector genome copies kg-1 (Cohort 1, n = 3) or 7.41 × 1013 vector gene copies kg-1 (Cohort 2, n = 3). Primary endpoint was safety, and secondary endpoint was change in SGCB expression in skeletal muscle from baseline to Day 60. We report interim Year 2 results (trial ongoing). The most frequent treatment-related adverse events were vomiting (four of six patients) and gamma-glutamyl transferase increase (three of six patients). Serious adverse events resolved with standard therapies. Robust SGCB expression was observed: Day 60 mean (s.d.) percentage of normal expression 36.2% (2.7%) in Cohort 1 and 62.1% (8.7%) in Cohort 2. Post hoc exploratory analysis showed preliminary motor improvements using the North Star Assessment for Limb-girdle Type Muscular Dystrophies maintained through Year 2. The 2-year safety and efficacy of bidridistrogene xeboparvovec support clinical development advancement. Further studies are necessary to confirm the long-term safety and efficacy of this gene therapy. ClinicalTrials.gov registration: NCT03652259 .


Assuntos
Distrofia Muscular do Cíngulo dos Membros , Sarcoglicanopatias , Humanos , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Distrofia Muscular do Cíngulo dos Membros/terapia , Sarcoglicanopatias/genética , Sarcoglicanopatias/metabolismo , Sarcoglicanopatias/terapia , Músculo Esquelético/metabolismo , Terapia Genética/efeitos adversos , Terapia Genética/métodos
2.
Int J Mol Sci ; 24(17)2023 Aug 31.
Artigo em Inglês | MEDLINE | ID: mdl-37686363

RESUMO

Dysferlinopathy treatment is an active area of investigation. Gene therapy is one potential approach. We studied muscle regeneration and inflammatory response after injection of an AAV-9 with a codon-optimized DYSF gene. A dual-vector system AAV.DYSF.OVERLAP with overlapping DYSF cDNA sequences was generated. Two AAV vectors were separately assembled by a standard triple-transfection protocol from plasmids carrying parts of the DYSF gene. Artificial myoblasts from dysferlin-deficient fibroblasts were obtained by MyoD overexpression. RT-PCR and Western blot were used for RNA and protein detection in vitro. A dysferlinopathy murine model (Bla/J) was used for in vivo studies. Histological assay, morphometry, and IHC were used for the muscle tissue analysis. Dysferlin was detected in vitro and in vivo at subphysiological levels. RT-PCR and Western Blot detected dysferlin mRNA and protein in AAV.DYSF.OVERLAP-transduced cells, and mRNA reached a 7-fold elevated level compared to the reference gene (GAPDH). In vivo, the experimental group showed intermediate median values for the proportion of necrotic muscle fibers, muscle fibers with internalized nuclei, and cross-sectional area of muscle fibers compared to the same parameters in the control groups of WT and Bla/J mice, although the differences were not statistically significant. The inverse relationship between the dosage and the severity of inflammatory changes in the muscles may be attributed to the decrease in the number of necrotic fibers. The share of transduced myofibers reached almost 35% in the group with the highest dose. The use of two-vector systems based on AAV is justified in terms of therapeutic efficacy. The expression of dysferlin at a subphysiological level, within a short observation period, is capable of inducing the restoration of muscle tissue structure, reducing inflammatory activity, and mitigating necrotic processes. Further research is needed to provide a more detailed assessment of the impact of the transgene and viral vector on the inflammatory component, including longer observation periods.


Assuntos
Dependovirus , Distrofia Muscular do Cíngulo dos Membros , Animais , Camundongos , Dependovirus/genética , Disferlina/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Códon , Fibras Musculares Esqueléticas , RNA Mensageiro
3.
Bull Exp Biol Med ; 174(6): 768-773, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37160600

RESUMO

We studied the effects of a dual-vector DYSF gene delivery system based on adeno-associated virus serotype 9 capsids on pathological manifestations of dysferlinopathy in skeletal muscles of Bla/J mice lacking DYSF expression. The mice received intravenous injection of 3×1013 genomic copies of the virus containing the dual-vector system. M. gastrocnemius, m. psoas major, m. vastus lateralis, and m. gluteus superficialis were isolated for histological examination in 3, 6, and 12 weeks after treatment. Healthy wild-type (C57BL/6) mice served as positive control and were sacrificed 3 weeks after injection of 150 µl of 0.9% NaCl into the caudal vein. To detect dysferlin in muscle cryosections, immunohistochemical analysis with diagnostic antibodies was performed; paraffin sections were stained with hematoxylin and eosin for morphometric analysis. After administration of gene-therapeutic constructs, muscle fibers with membrane or cytoplasmic dysferlin location were detected in all examined muscles. The proportion of necrotic muscle fibers decreased, the number of muscle fibers with central location of the nucleus increased, and the mean cross-section area of the muscle fibers decreased.


Assuntos
Músculo Esquelético , Distrofia Muscular do Cíngulo dos Membros , Camundongos , Animais , Disferlina/genética , Disferlina/metabolismo , Camundongos Endogâmicos C57BL , Músculo Esquelético/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Fibras Musculares Esqueléticas/metabolismo , Técnicas de Transferência de Genes
4.
Ann Clin Transl Neurol ; 10(5): 686-695, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37026610

RESUMO

Valosin-containing protein (VCP)-associated multisystem proteinopathy (MSP) is a rare genetic disorder with abnormalities in the autophagy pathway leading to various combinations of myopathy, bone diseases, and neurodegeneration. Ninety percent of patients with VCP-associated MSP have myopathy, but there is no consensus-based guideline. The goal of this working group was to develop a best practice set of provisional recommendations for VCP myopathy which can be easily implemented across the globe. As an initiative by Cure VCP Disease Inc., a patient advocacy organization, an online survey was initially conducted to identify the practice gaps in VCP myopathy. All prior published literature on VCP myopathy was reviewed to better understand the different aspects of management of VCP myopathy, and several working group sessions were conducted involving international experts to develop this provisional recommendation. VCP myopathy has a heterogeneous clinical phenotype and should be considered in patients with limb-girdle muscular dystrophy phenotype, or any myopathy with an autosomal dominant pattern of inheritance. Genetic testing is the only definitive way to diagnose VCP myopathy, and single-variant testing in the case of a known familial VCP variant, or multi-gene panel sequencing in undifferentiated cases can be considered. Muscle biopsy is important in cases of diagnostic uncertainty or lack of a definitive pathogenic genetic variant since rimmed vacuoles (present in ~40% cases) are considered a hallmark of VCP myopathy. Electrodiagnostic studies and magnetic resonance imaging can also help rule out disease mimics. Standardized management of VCP myopathy will optimize patient care and help future research initiatives.


Assuntos
Doenças Musculares , Distrofia Muscular do Cíngulo dos Membros , Deficiências na Proteostase , Humanos , Proteína com Valosina/genética , Doenças Musculares/diagnóstico , Doenças Musculares/genética , Doenças Musculares/terapia , Distrofia Muscular do Cíngulo dos Membros/diagnóstico , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Fenótipo
5.
J Clin Invest ; 132(1)2022 01 04.
Artigo em Inglês | MEDLINE | ID: mdl-34981776

RESUMO

Efficient sarcolemmal repair is required for muscle cell survival, with deficits in this process leading to muscle degeneration. Lack of the sarcolemmal protein dysferlin impairs sarcolemmal repair by reducing secretion of the enzyme acid sphingomyelinase (ASM), and causes limb girdle muscular dystrophy 2B (LGMD2B). The large size of the dysferlin gene poses a challenge for LGMD2B gene therapy efforts aimed at restoring dysferlin expression in skeletal muscle fibers. Here, we present an alternative gene therapy approach targeting reduced ASM secretion, the consequence of dysferlin deficit. We showed that the bulk endocytic ability is compromised in LGMD2B patient cells, which was addressed by extracellularly treating cells with ASM. Expression of secreted human ASM (hASM) using a liver-specific adeno-associated virus (AAV) vector restored membrane repair capacity of patient cells to healthy levels. A single in vivo dose of hASM-AAV in the LGMD2B mouse model restored myofiber repair capacity, enabling efficient recovery of myofibers from focal or lengthening contraction-induced injury. hASM-AAV treatment was safe, attenuated fibro-fatty muscle degeneration, increased myofiber size, and restored muscle strength, similar to dysferlin gene therapy. These findings elucidate the role of ASM in dysferlin-mediated plasma membrane repair and to our knowledge offer the first non-muscle-targeted gene therapy for LGMD2B.


Assuntos
Dependovirus , Terapia Genética , Vetores Genéticos , Fígado/enzimologia , Distrofia Muscular do Cíngulo dos Membros , Esfingomielina Fosfodiesterase , Animais , Linhagem Celular Transformada , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Mutantes , Distrofia Muscular do Cíngulo dos Membros/enzimologia , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Esfingomielina Fosfodiesterase/biossíntese , Esfingomielina Fosfodiesterase/genética
6.
Hum Gene Ther ; 32(7-8): 390-404, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33349138

RESUMO

Limb-girdle muscular dystrophy type 2D/R3 (LGMD2D/R3) is a progressive muscular dystrophy that manifests with muscle weakness, respiratory abnormalities, and in rare cases cardiomyopathy. LGMD2D/R3 is caused by mutations in the SGCA gene resulting in loss of protein and concomitant loss of some or all components of the dystrophin-associated glycoprotein complex. The sgca-null (sgca-/-) mouse recapitulates the clinical phenotype of patients with LGMD2D/R3, including dystrophic features such as muscle necrosis and fibrosis, elevated serum creatine kinase (CK), and reduction in the generation of absolute muscle force and locomotor activity. Thus, sgca-/- mice provide a relevant model to test the safety and efficacy of gene transfer. We designed a self-complementary AAVrh74 vector containing a codon-optimized full-length human SGCA (hSGCA) transgene driven by a muscle-specific promoter, shortened muscle creatine kinase (tMCK). In this report, we test the efficacy and safety of scAAVrh74.tMCK.hSGCA in sgca-/- mice using a dose-escalation design to evaluate a single systemic injection of 1.0 × 1012, 3.0 × 1012, and 6.0 × 1012 vg total dose compared with vehicle-treatment and wild-type mice. In sgca-/- mice, treatment with scAAVrh74.tMCK.hSGCA resulted in robust expression of α-sarcoglycan protein at the sarcolemma membrane in skeletal muscle at all doses tested. In addition, scAAVrh74.tMCK.hSGCA was effective in improving the histopathology of limb and diaphragm muscle of sgca-/- mice, as indicated by reductions in fibrosis, central nucleation, and normalization of myofiber size. These molecular changes were concomitant with significant increases in specific force generation in the diaphragm and tibialis anterior muscle, protection against eccentric force loss, and reduction in serum CK. Locomotor activity was improved at all doses of vector-treated compared with vehicle-treated sgca-/- mice. Lastly, vector toxicity was not detected in a serum chemistry panel and by gross necropsy. Collectively, these findings provide support for a systemic delivery of scAAVrh74.tMCK.hSGCA in a clinical setting for the treatment of LGMD2D/R3.


Assuntos
Distrofia Muscular do Cíngulo dos Membros , Sarcoglicanopatias , Animais , Terapia Genética , Humanos , Camundongos , Músculo Esquelético , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Sarcoglicanopatias/genética , Sarcoglicanopatias/terapia , Sarcoglicanas/genética
7.
Skelet Muscle ; 10(1): 22, 2020 07 29.
Artigo em Inglês | MEDLINE | ID: mdl-32727611

RESUMO

The limb-girdle muscular dystrophies (LGMDs) are a genetically pleiomorphic class of inherited muscle diseases that are known to share phenotypic features. Selected LGMD genetic subtypes have been studied extensively in affected humans and various animal models. In some cases, these investigations have led to human clinical trials of potential disease-modifying therapies, including gene replacement strategies for individual subtypes using adeno-associated virus (AAV) vectors. The cellular localizations of most proteins associated with LGMD have been determined. However, the functions of these proteins are less uniformly characterized, thus limiting our knowledge of potential common disease mechanisms across subtype boundaries. Correspondingly, broad therapeutic strategies that could each target multiple LGMD subtypes remain less developed. We believe that three major "functional clusters" of subcellular activities relevant to LGMD merit further investigation. The best known of these is the glycosylation modifications associated with the dystroglycan complex. The other two, mechanical signaling and mitochondrial dysfunction, have been studied less systematically but are just as promising with respect to the identification of significant mechanistic subgroups of LGMD. A deeper understanding of these disease pathways could yield a new generation of precision therapies that would each be expected to treat a broader range of LGMD patients than a single subtype, thus expanding the scope of the molecular medicines that may be developed for this complex array of muscular dystrophies.


Assuntos
Distrofia Muscular do Cíngulo dos Membros/genética , Animais , Terapia Genética/métodos , Humanos , Mitocôndrias Musculares/metabolismo , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Distrofia Muscular do Cíngulo dos Membros/terapia
8.
Neurol Clin ; 38(3): 493-504, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32703463

RESUMO

The limb girdle muscular dystrophies (LGMDs) are genetic muscle diseases with primary skeletal muscle involvement in persons with the ability to walk independently at some point in the disease course. They usually have increased creatine kinase levels along with patterns of fatty and fibrous deposition on muscle imaging and/or dystrophic features on muscle biopsy. Distinctive clinical features provide valuable diagnostic clues to the diagnosis and sometimes treatment of these disorders. The advent of gene and cell-based therapies; gene replacement, editing, and modulation; along with stem cell and small molecule therapies may significantly ameliorate clinical severity in the LGMDs.


Assuntos
Músculo Esquelético/diagnóstico por imagem , Distrofia Muscular do Cíngulo dos Membros/diagnóstico por imagem , Distrofia Muscular do Cíngulo dos Membros/genética , Adulto , Progressão da Doença , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Distrofia Muscular do Cíngulo dos Membros/terapia
9.
Skelet Muscle ; 10(1): 10, 2020 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-32321586

RESUMO

BACKGROUND: Defects in α-dystroglycan (DG) glycosylation characterize a group of muscular dystrophies known as dystroglycanopathies. One of the key effectors in the α-DG glycosylation pathway is the glycosyltransferase fukutin-related protein (FKRP). Mutations in FKRP lead to a large spectrum of muscular dystrophies, including limb girdle muscular dystrophy 2I (LGMD2I). It remains unknown whether stem cell transplantation can promote muscle regeneration and ameliorate the muscle wasting phenotype associated with FKRP mutations. RESULTS: Here we transplanted murine and human pluripotent stem cell-derived myogenic progenitors into a novel immunodeficient FKRP-mutant mouse model by intra-muscular injection. Upon both mouse and human cell transplantation, we observe the presence of donor-derived myofibers even in absence of pre-injury, and the rescue of α-DG functional glycosylation, as shown by IIH6 immunoreactivity. The presence of donor-derived cells expressing Pax7 under the basal lamina is indicative of satellite cell engraftment, and therefore, long-term repopulation potential. Functional assays performed in the mouse-to-mouse cohort revealed enhanced specific force in transplanted muscles compared to PBS-injected controls. CONCLUSIONS: Altogether, our data demonstrate for the first time the suitability of a cell-based therapeutic approach to improve the muscle phenotype of dystrophic FKRP-mutant mice.


Assuntos
Terapia Genética/métodos , Fibras Musculares Esqueléticas/citologia , Distrofia Muscular do Cíngulo dos Membros/terapia , Pentosiltransferases/genética , Transplante de Células-Tronco/métodos , Animais , Diferenciação Celular , Células Cultivadas , Distroglicanas/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos SCID , Fibras Musculares Esqueléticas/metabolismo , Distrofia Muscular do Cíngulo dos Membros/genética , Fator de Transcrição PAX7/genética , Fator de Transcrição PAX7/metabolismo , Pentosiltransferases/metabolismo , Células-Tronco Pluripotentes/citologia , Células-Tronco Pluripotentes/metabolismo
10.
Curr Gene Ther ; 19(6): 386-394, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32067617

RESUMO

The Limb-Girdle Muscular Dystrophies (LGMD) are genetically heterogeneous disorders, responsible for muscle wasting and severe form of dystrophies. Despite the critical developments in the insight and information of pathomechanisms of limb-girdle muscular dystrophy, any definitive treatments do not exist, and current strategies are only based on the improvement of the signs of disorder and to enhance the life quality without resolving an underlying cause. There is a crucial relationship between pharmacological therapy and different consequences; therefore, other treatment strategies will be required. New approaches, such as gene replacement, gene transfer, exon skipping, siRNA knockdown, and anti-myostatin therapy, which can target specific cellular or molecular mechanism of LGMD, could be a promising avenue for the treatment. Recently, genome engineering strategies with a focus on molecular tools such as CRISPR-Cas9 are used to different types of neuromuscular disorders and show the highest potential for clinical translation of these therapies. Thus, recent advancements and challenges in the field will be reviewed in this paper.


Assuntos
Terapia Genética , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Animais , Sistemas CRISPR-Cas , Modelos Animais de Doenças , Éxons , Edição de Genes , Humanos , Células-Tronco Pluripotentes Induzidas , Células-Tronco Mesenquimais , Camundongos , RNA Interferente Pequeno , Transplantes
11.
Med Sci (Paris) ; 36 Hors série n° 2: 22-27, 2020 Dec.
Artigo em Francês | MEDLINE | ID: mdl-33427632

RESUMO

Sarcoglycanopathies are the third most common cause of autosomal recessive limb girdle muscular dystrophies (LGMD). They are the result of a deficiency in one of the sarcoglycans a, b, g, or d. The usual clinical presentation is that of a symmetrical involvement of the muscles of the pelvic and scapular girdles as well as of the trunk, associated with more or less severe cardio-respiratory impairment and a marked increase of serum CK levels. The first symptoms appear during the first decade, the loss of ambulation occurring often during the second decade. Lesions observed on the muscle biopsy are dystrophic. This is associated with a decrease or an absence of immunostaining of the sarcoglycan corresponding to the mutated gene and, to a lesser degree, of the other three sarcoglycans. Many mutations have been reported in the four incriminated genes and some of them are prevalent in certain populations. To date, there is no curative treatment, which does not prevent the development of many clinical trials, especially in gene therapy.


TITLE: Les sarcoglycanopathies - État des lieux et perspectives thérapeutiques. ABSTRACT: Les sarcoglycanopathies font partie des dystrophies musculaires des ceintures (LGMD) autosomiques récessives et représentent la troisième cause la plus fréquente d'entre elles. Elles sont consécutives à un déficit d'un des sarcoglycanes α, ß, γ, ou δ. La présentation clinique habituelle est celle d'une atteinte symétrique des muscles des ceintures pelvienne et scapulaire ainsi que du tronc, associée à une atteinte cardiorespiratoire plus ou moins sévère et une élévation franche des créatine-phospho-kinases (CPK). Les premiers symptômes apparaissent au cours de la première décennie, la perte de la marche survenant souvent au cours de la deuxième décennie. Les lésions sont de type dystrophique sur la biopsie musculaire. Il s'y associe une diminution ou une absence d'immunomarquage du sarcoglycane correspondant au gène muté, et dans une moindre mesure des trois autres sarcoglycanes associés. De nombreuses mutations ont été rapportées dans les quatre gènes impliqués et quelques-unes d'entre elles sont prépondérantes dans certaines populations. à ce jour, il n'existe pas de traitement curatif ce qui n'empêche pas de voir se développer de nombreux essais cliniques, notamment en thérapie génique.


Assuntos
Sarcoglicanopatias/terapia , Progressão da Doença , Humanos , Distrofia Muscular do Cíngulo dos Membros/classificação , Distrofia Muscular do Cíngulo dos Membros/diagnóstico , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Mutação , Neurologia/métodos , Neurologia/tendências , Sarcoglicanopatias/diagnóstico , Sarcoglicanopatias/epidemiologia , Sarcoglicanopatias/genética , Terapias em Estudo/métodos , Terapias em Estudo/tendências
12.
Med Sci (Paris) ; 36 Hors série n° 2: 28-33, 2020 Dec.
Artigo em Francês | MEDLINE | ID: mdl-33427633

RESUMO

Mutations in the FKRP gene encoding the fukutin-related protein (FKRP) cause a wide spectrum of myopathies, ranging from severe forms of congenital muscular dystrophies associated with structural abnormalities of the central nervous system, to exertional myalgia or asymptomatic hyperCKemia, and to a form of limb girdle muscular dystrophy, LGMD-R9, (ex-LGMD-2I). LGMD-R9 is characterized by a proximal girdle deficit predominantly in the lower limbs to start with, with respiratory and cardiac damage that may affect the vital prognosis. Serum CK levels are markedly elevated and, on muscle biopsy, is detected a dystrophic formula associated with a reduction in the glycosylation of α-dystroglycan by immunostains and immunoblotting. Muscle MRI typically shows damage to proximal muscles (iliopsoas, adductors, gluteus maximus, quadriceps) with relative preservation of the muscles of the anterior compartment of the thighs (gracilis and sartorius). Genetic analysis, by specific sequencing of the FKRP gene or of a panel grouping together all the genes involved in the glycosylation of α-dystroglycan, or a larger panel of genes, generally confirms the diagnosis, the most frequent mutation being the missense p.(Leu276Ile). Currently, treatment of LGMD-R9 is symptomatic, requiring a multidisciplinary approach. A prospective study of the natural history of the disease is currently underway in Europe (GNT-015-FKRP). New therapeutic approaches are envisaged, such as gene therapy mediated by vectors derived from the adeno-associated virus (AAV). This is effective in animal models, allowing correction of defects in the glycosylation of alpha-dystroglycan and an increase in its binding capacity to the extracellular matrix. At the same time, preclinical studies have shown, in an animal model, the efficacy of ribitol, an alcohol pentose found in natural compounds, which has led to a phase I trial whose clinical development is underway.


TITLE: La dystrophie musculaire des ceintures de type R9 liée au gène FKRP - État des lieux et perspectives thérapeutiques. ABSTRACT: Les mutations du gène FKRP codant la fukutin-related protein (FKRP) sont à l'origine d'un large éventail de myopathies allant de formes sévères de dystrophies musculaires congénitales associées à des anomalies structurales du système nerveux central, jusqu'à des tableaux de myalgies à l'effort ou d'hyperCKémie asymptomatique, en passant par une forme de dystrophie musculaire des ceintures, la LGMD-R9 (ex-LGMD-2I), pour limb girdle muscular dystrophy récessive de type R9. La LGMD-R9 se caractérise par un déficit proximal des ceintures prédominant initialement aux membres inférieurs, avec une atteinte respiratoire et cardiaque pouvant conditionner le pronostic vital. Le taux sérique de CPK est nettement élevé et s'accompagne, sur la biopsie musculaire, d'une formule dystrophique associée à une réduction de la glycosylation de l'α-dystroglycane visible en immunomarquage et par immunoblot. L'IRM musculaire montre typiquement une atteinte des muscles proximaux (iliopsoas, adducteurs, grands fessiers, quadriceps) avec une relative préservation des muscles de la loge antérieure des cuisses (gracilis et sartorius). L'analyse génétique, par séquençage spécifique du gène FKRP ou d'un panel regroupant l'ensemble des gènes impliqués dans la glycosylation de l'α-dystroglycane, ou bien d'un panel plus large de gènes, confirme généralement le diagnostic, la mutation la plus fréquente étant le faux-sens p.(Leu276Ile). Actuellement, le traitement de la LGMD-R9 est symptomatique, requérant une approche pluridisciplinaire. Une étude prospective d'histoire naturelle de la maladie est en cours en Europe (GNT-015-FKRP). Des approches thérapeutiques inédites sont envisagées, telles que la thérapie génique médiée par des vecteurs dérivés du virus adéno-associé (AAV). Celle-ci est efficace dans les modèles animaux, permettant une correction des défauts de glycosylation de l'a-dystroglycane et une augmentation de sa capacité de liaison à la matrice extracellulaire. En parallèle, des études précliniques ont montré, dans un modèle animal, l'efficacité du ribitol, un pentose alcool retrouvé dans des composés naturels, ce qui a conduit à un essai de phase I dont le développement clinique est en cours.


Assuntos
Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Pentosiltransferases/genética , Animais , Diagnóstico Diferencial , Humanos , Distrofia Muscular do Cíngulo dos Membros/diagnóstico , Distrofia Muscular do Cíngulo dos Membros/epidemiologia , Mutação de Sentido Incorreto , Ribitol/uso terapêutico , Terapias em Estudo/métodos , Terapias em Estudo/tendências
13.
Sci Transl Med ; 11(520)2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31776291

RESUMO

Limb-girdle muscular dystrophy type 2A (LGMD2A or LGMDR1) is a neuromuscular disorder caused by mutations in the calpain 3 gene (CAPN3). Previous experiments using adeno-associated viral (AAV) vector-mediated calpain 3 gene transfer in mice indicated cardiac toxicity associated with the ectopic expression of the calpain 3 transgene. Here, we performed a preliminary dose study in a severe double-knockout mouse model deficient in calpain 3 and dysferlin. We evaluated safety and biodistribution of AAV9-desmin-hCAPN3 vector administration to nonhuman primates (NHPs) with a dose of 3 × 1013 viral genomes/kg. Vector administration did not lead to observable adverse effects or to detectable toxicity in NHP. Of note, the transgene expression did not produce any abnormal changes in cardiac morphology or function of injected animals while reaching therapeutic expression in skeletal muscle. Additional investigation on the underlying causes of cardiac toxicity observed after gene transfer in mice and the role of titin in this phenomenon suggest species-specific titin splicing. Mice have a reduced capacity for buffering calpain 3 activity compared to NHPs and humans. Our studies highlight a complex interplay between calpain 3 and titin binding sites and demonstrate an effective and safe profile for systemic calpain 3 vector delivery in NHP, providing critical support for the clinical potential of calpain 3 gene therapy in humans.


Assuntos
Calpaína/genética , Calpaína/uso terapêutico , Cardiotoxicidade/etiologia , Conectina/genética , Terapia Genética/efeitos adversos , Proteínas Musculares/genética , Proteínas Musculares/uso terapêutico , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Splicing de RNA/genética , Animais , Sítios de Ligação , Biomarcadores/sangue , Cardiotoxicidade/sangue , Conectina/química , Dependovirus/genética , Disferlina/deficiência , Disferlina/metabolismo , Estabilidade Enzimática , Regulação da Expressão Gênica , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , Músculo Esquelético/metabolismo , Distrofia Muscular do Cíngulo dos Membros/sangue , Distrofia Muscular do Cíngulo dos Membros/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Primatas , Domínios Proteicos , Proteólise , Especificidade da Espécie , Distribuição Tecidual , Transgenes
14.
Mol Ther ; 27(12): 2147-2157, 2019 12 04.
Artigo em Inglês | MEDLINE | ID: mdl-31501033

RESUMO

Limb girdle muscular dystrophy type 2A (LGMD2A), caused by mutations in the Calpain 3 (CAPN3) gene, is an incurable autosomal recessive disorder that results in muscle wasting and loss of ambulation. To test the feasibility of an autologous induced pluripotent stem cell (iPSC)-based therapy for LGMD2A, here we applied CRISPR-Cas9-mediated genome editing to iPSCs from three LGMD2A patients to enable correction of mutations in the CAPN3 gene. Using a gene knockin approach, we genome edited iPSCs carrying three different CAPN3 mutations, and we demonstrated the rescue of CAPN3 protein in myotube derivatives in vitro. Transplantation of gene-corrected LGMD2A myogenic progenitors in a novel mouse model combining immunodeficiency and a lack of CAPN3 resulted in muscle engraftment and rescue of the CAPN3 mRNA. Thus, we provide here proof of concept for the integration of genome editing and iPSC technologies to develop a novel autologous cell therapy for LGMD2A.


Assuntos
Calpaína/fisiologia , Terapia Baseada em Transplante de Células e Tecidos/métodos , Células-Tronco Pluripotentes Induzidas/citologia , Proteínas Musculares/fisiologia , Músculo Esquelético/metabolismo , Distrofia Muscular do Cíngulo dos Membros/terapia , Mutação , Animais , Células Cultivadas , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patologia , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/patologia , Transplante Autólogo
15.
Hum Gene Ther ; 30(7): 794-801, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30838895

RESUMO

In a previous limb-girdle muscular dystrophy type 2D (LGMD2D) clinical trial, robust alpha-sarcoglycan gene expression was confirmed following intramuscular gene (SGCA) transfer. This paved the way for first-in-human isolated limb infusion (ILI) gene transfer trial to the lower limbs. Delivery of scAAVrh74.tMCK.hSGCA via an intravascular route through the femoral artery predicted improved ambulation. This method was initially chosen to avoid safety concerns required for large systemic vascular delivery viral loads. ILI methods were adopted from the extensive chemotherapy experience for treatment of malignancies confined to the extremities. Six LGMD2D subjects were enrolled in a dose-ascending open-label clinical trial. Safety of the procedure was initially assessed in the single limb of a non-ambulant affected adult at a dose of 1 × 1012 vg/kg. Subsequently, ambulatory children (aged 8-13 years) were enrolled and dosed bilaterally with either 1 × 1012 vg/kg/limb or 3 × 1012 vg/kg/limb. The six-minute walk test (6MWT) served as the primary clinical outcome; secondary outcomes included muscle strength (maximum voluntary isometric force testing) and SGCA expression at 6 months. All ambulatory participants except one had pre- and post-treatment muscle biopsies. All four subjects biopsied had confirmed SGCA gene delivery by immunofluorescence, Western blot analysis (14-25% of normal), and vector genome copies (5.4 × 103-7.7 × 104 vg/µg). Muscle strength in the knee extensors (assessed by force generation in kilograms) showed improvement in two subjects that correlated with an increase in fiber diameter post gene delivery. Six-minute walk times decreased or remained the same. Vascular delivery of AAVrh74.tMCK.hSGCA was effective at producing SGCA protein at low doses that correlated with vector copies and local functional improvement restricted to targeted muscles. Future trials will focus on systemic administration to enable targeting of proximal muscles to maximize clinical benefit.


Assuntos
Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Sarcoglicanopatias/genética , Transgenes , Animais , Biomarcadores , Criança , Modelos Animais de Doenças , Feminino , Expressão Gênica , Vetores Genéticos/administração & dosagem , Humanos , Injeções Intramusculares , Masculino , Pessoa de Meia-Idade , Distrofia Muscular do Cíngulo dos Membros/fisiopatologia , Transdução Genética , Resultado do Tratamento
17.
Pract Neurol ; 18(3): 201-210, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29472383

RESUMO

The expansion of the spectrum of limb girdle muscular dystrophies (LGMDs) in recent years means that neurologists need to be familiar with the clinical clues that can help with their diagnosis. The LGMDs comprise a group of genetic myopathies that manifest as chronic progressive weakness of hip and shoulder girdles. Their inheritance is either autosomal dominant (LGMD1) or autosomal recessive (LGMD2). Their prevalence varies in different regions of the world; certain ethnic groups have documented founder mutations and this knowledge can facilitate the diagnosis. The clinical approach to LGMDs uses the age at onset, genetic transmission and clinical patterns of muscular weakness. Helpful clinical features that help to differentiate the various subtypes include: predominant upper girdle weakness, disproportionate respiratory muscle involvement, distal weakness, hip adductor weakness, 'biceps lump' and 'diamond on quadriceps' sign, calf hypertrophy, contractures and cardiac involvement. Almost half of patients with LGMD have such clinical clues. Investigations such as serum creatine kinase, electrophysiology, muscle biopsy and genetic studies can complement the clinical examination. In this review, we discuss diagnostic clinical pointers and comment on the differential diagnosis and relevant investigations, using illustrative case studies.


Assuntos
Distrofia Muscular do Cíngulo dos Membros , Adulto , Idade de Início , Criança , Creatina Quinase/sangue , Eletrofisiologia , Humanos , Masculino , Debilidade Muscular/etiologia , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/diagnóstico , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Adulto Jovem
18.
Hum Gene Ther ; 29(7): 749-762, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-28707952

RESUMO

Dysferlinopathies comprise a family of disorders caused by mutations in the dysferlin (DYSF) gene, leading to a progressive dystrophy characterized by chronic muscle fiber loss, fat replacement, and fibrosis. To correct the underlying histopathology and function, expression of full-length DYSF is required. Dual adeno-associated virus vectors have been developed, defined by a region of homology, to serve as a substrate for reconstitution of the full 6.5 kb dysferlin cDNA. Previous work studied the efficacy of this treatment through intramuscular and regional delivery routes. To maximize clinical efficacy, dysferlin-deficient mice were treated systemically to target all muscles through the vasculature for efficacy and safety studies. Mice were evaluated at multiple time points between 4 and 13 months post treatment for dysferlin expression and functional improvement using magnetic resonance imaging and magnetic resonance spectroscopy and membrane repair. A systemic dose of 6 × 1012 vector genomes resulted in widespread gene expression in the muscles. Treated muscles showed a significant decrease in central nucleation, collagen deposition, and improvement of membrane repair to wild-type levels. Treated gluteus muscles were significantly improved compared to placebo-treated muscles and were equivalent to wild type in volume, intra- and extramyocellular lipid accumulation, and fat percentage using magnetic resonance imaging and magnetic resonance spectroscopy. Dual-vector treatment allows for production of full-length functional dysferlin with no toxicity. This confirms previous safety data and validates translation of systemic gene delivery for dysferlinopathy patients.


Assuntos
DNA Complementar/administração & dosagem , Disferlina/genética , Terapia Genética , Distrofia Muscular do Cíngulo dos Membros/terapia , Animais , DNA Complementar/genética , Dependovirus/genética , Modelos Animais de Doenças , Disferlina/administração & dosagem , Regulação da Expressão Gênica , Vetores Genéticos/uso terapêutico , Humanos , Masculino , Camundongos , Músculo Esquelético , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/patologia , Mutação
19.
Mol Ther ; 25(9): 2150-2162, 2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28629822

RESUMO

Dysferlinopathy is an autosomal recessive muscular dystrophy characterized by the progressive loss of motility that is caused by mutations throughout the DYSF gene. There are currently no approved therapies that ameliorate or reverse dysferlinopathy. Gene delivery using adeno-associated vectors (AAVs) is a leading therapeutic strategy for genetic diseases; however, the large size of dysferlin cDNA (6.2 kB) precludes packaging into a single AAV capsid. Therefore, using 3D structural modeling and hypothesizing dysferlin C2 domain redundancy, a 30% smaller, dysferlin-like molecule amenable to single AAV vector packaging was engineered (termed Nano-Dysferlin). The intracellular distribution of Nano-Dysferlin was similar to wild-type dysferlin and neither demonstrated toxicity when overexpressed in dysferlin-deficient patient myoblasts. Intramuscular injection of AAV-Nano-Dysferlin in young dysferlin-deficient mice significantly improved muscle integrity and decreased muscle turnover 3 weeks after treatment, as determined by Evans blue dye uptake and central nucleated fibers, respectively. Systemically administered AAV-Nano-Dysferlin to young adult dysferlin-deficient mice restored motor function and improved muscle integrity nearly 8 months after a single injection. These preclinical data are the first report of a smaller dysferlin variant tailored for AAV single particle delivery that restores motor function and, therefore, represents an attractive candidate for the treatment of dysferlinopathy.


Assuntos
Desenho de Fármacos , Disferlina/química , Disferlina/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Animais , Dependovirus/genética , Modelos Animais de Doenças , Disferlina/metabolismo , Ordem dos Genes , Técnicas de Transferência de Genes , Terapia Genética , Vetores Genéticos/administração & dosagem , Vetores Genéticos/genética , Injeções Intramusculares , Camundongos , Atividade Motora/genética , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Músculo Esquelético/fisiopatologia , Distrofia Muscular do Cíngulo dos Membros/metabolismo , Distrofia Muscular do Cíngulo dos Membros/terapia , Regiões Promotoras Genéticas , Domínios Proteicos , Relação Estrutura-Atividade , Transdução Genética
20.
Neuromuscul Disord ; 27(8): 738-741, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28602176

RESUMO

Mutations in calpain-3 cause limb girdle muscular dystrophy 2A. Biopsy pathology is typically dystrophic, sometimes characterized by frequent lobulated fibres. More recently calpain mutations have been shown in association with eosinophilic myositis, suggesting that calpain mutations may render muscle susceptible to inflammatory change. We present the case of a 33-year old female with mild proximal muscle weakness and high CK levels (6698 IU/L at presentation). Muscle biopsy showed clusters of fibre necrosis associated with very dense macrophage infiltrates and small numbers of lymphocytes, raising the possibility of an inflammatory myopathy. No eosinophils were observed. Immunosuppressive treatment was started without clinical improvement. MRI demonstrated bilateral fatty replacement in posterior thigh and calf muscles. Western blot results prompted Sanger sequencing of the calpain-3 gene revealing compound heterozygous mutations c.643_663del and c.1746-20C>G. Our case widens the myopathological spectrum of calpainopathies to include focal macrophage rich inflammatory change.


Assuntos
Calpaína/genética , Macrófagos/patologia , Proteínas Musculares/genética , Distrofia Muscular do Cíngulo dos Membros/genética , Distrofia Muscular do Cíngulo dos Membros/patologia , Adulto , Calpaína/metabolismo , Feminino , Humanos , Terapia de Imunossupressão , Macrófagos/imunologia , Proteínas Musculares/metabolismo , Músculo Esquelético/diagnóstico por imagem , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Distrofia Muscular do Cíngulo dos Membros/fisiopatologia , Distrofia Muscular do Cíngulo dos Membros/terapia , Mutação , Necrose
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA