Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
1.
JCI Insight ; 9(11)2024 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-38753465

RESUMO

Glycogen storage disease type III (GSDIII) is a rare metabolic disorder due to glycogen debranching enzyme (GDE) deficiency. Reduced GDE activity leads to pathological glycogen accumulation responsible for impaired hepatic metabolism and muscle weakness. To date, there is no curative treatment for GSDIII. We previously reported that 2 distinct dual AAV vectors encoding for GDE were needed to correct liver and muscle in a GSDIII mouse model. Here, we evaluated the efficacy of rapamycin in combination with AAV gene therapy. Simultaneous treatment with rapamycin and a potentially novel dual AAV vector expressing GDE in the liver and muscle resulted in a synergic effect demonstrated at biochemical and functional levels. Transcriptomic analysis confirmed synergy and suggested a putative mechanism based on the correction of lysosomal impairment. In GSDIII mice livers, dual AAV gene therapy combined with rapamycin reduced the effect of the immune response to AAV observed in this disease model. These data provide proof of concept of an approach exploiting the combination of gene therapy and rapamycin to improve efficacy and safety and to support clinical translation.


Assuntos
Dependovirus , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos , Fígado , Sirolimo , Animais , Sirolimo/farmacologia , Sirolimo/uso terapêutico , Dependovirus/genética , Terapia Genética/métodos , Camundongos , Fígado/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/administração & dosagem , Músculo Esquelético/metabolismo , Fenótipo , Sistema da Enzima Desramificadora do Glicogênio/genética , Sistema da Enzima Desramificadora do Glicogênio/metabolismo , Humanos , Masculino
2.
J Clin Invest ; 134(2)2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38015640

RESUMO

Glycogen storage disease type III (GSDIII) is a rare inborn error of metabolism affecting liver, skeletal muscle, and heart due to mutations of the AGL gene encoding for the glycogen debranching enzyme (GDE). No curative treatment exists for GSDIII. The 4.6 kb GDE cDNA represents the major technical challenge toward the development of a single recombinant adeno-associated virus-derived (rAAV-derived) vector gene therapy strategy. Using information on GDE structure and molecular modeling, we generated multiple truncated GDEs. Among them, an N-terminal-truncated mutant, ΔNter2-GDE, had a similar efficacy in vivo compared with the full-size enzyme. A rAAV vector expressing ΔNter2-GDE allowed significant glycogen reduction in heart and muscle of Agl-/- mice 3 months after i.v. injection, as well as normalization of histology features and restoration of muscle strength. Similarly, glycogen accumulation and histological features were corrected in a recently generated Agl-/- rat model. Finally, transduction with rAAV vectors encoding ΔNter2-GDE corrected glycogen accumulation in an in vitro human skeletal muscle cellular model of GSDIII. In conclusion, our results demonstrated the ability of a single rAAV vector expressing a functional mini-GDE transgene to correct the muscle and heart phenotype in multiple models of GSDIII, supporting its clinical translation to patients with GSDIII.


Assuntos
Sistema da Enzima Desramificadora do Glicogênio , Doença de Depósito de Glicogênio Tipo III , Humanos , Camundongos , Ratos , Animais , Doença de Depósito de Glicogênio Tipo III/genética , Doença de Depósito de Glicogênio Tipo III/terapia , Sistema da Enzima Desramificadora do Glicogênio/genética , Músculo Esquelético/metabolismo , Glicogênio/metabolismo , Transgenes
3.
J Orthop Res ; 42(5): 1111-1121, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-37975418

RESUMO

Although the etiology of intervertebral disc degeneration is still unresolved, the nutrient paucity resulting from its avascular nature is suspected of triggering degenerative processes in its core: the nucleus pulposus (NP). While severe hypoxia has no significant effects on NP cells, the impact of glucose depletion, such as found in degenerated discs (0.2-1 mM), is still uncertain. Using a pertinent ex-vivo model representative of the unique disc microenvironment, the present study aimed, therefore, at determining the effects of "degenerated" (0.3 mM) glucose levels on bovine NP explant homeostasis. The effects of glucose depletion were evaluated on NP cell viability, apoptosis, phenotype, metabolism, senescence, extracellular matrix anabolism and catabolism, and inflammatory mediator production using fluorescent staining, RT-qPCR, (immuno)histology, ELISA, biochemical, and enzymatic assays. Compared to the "healthy" (2 mM) glucose condition, exposure to the degenerated glucose condition led to a rapid and extensive decrease in NP cell viability associated with increased apoptosis. Although the aggrecan and collagen-II gene expression was also downregulated, NP cell phenotype, and senescence, matrix catabolism, and inflammatory mediator production were not, or only slightly, affected by glucose depletion. The present study provided evidence for glucose depletion as an essential player in NP cell viability but also suggested that other microenvironment factor(s) may be involved in triggering the typical shift of NP cell phenotype observed during disc degeneration. The present study contributes new information for better understanding disc degeneration at the cellular-molecular levels and thus helps to develop relevant therapeutical strategies to counteract it.


Assuntos
Degeneração do Disco Intervertebral , Disco Intervertebral , Núcleo Pulposo , Animais , Bovinos , Núcleo Pulposo/metabolismo , Degeneração do Disco Intervertebral/patologia , Sobrevivência Celular , Glucose/metabolismo , Mediadores da Inflamação/metabolismo , Disco Intervertebral/patologia
4.
Stem Cell Res ; 72: 103214, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37769385

RESUMO

Glycogen storage disease type III (GSDIII) is an autosomal recessive disorder characterized by a deficiency of glycogen debranching enzyme (GDE) leading to cytosolic glycogen accumulation and inducing liver and muscle pathology. Skin fibroblasts from three GSDIII patients were reprogrammed into induced pluripotent stem cells (iPSCs) using non-integrated Sendai virus. All of the three lines exhibited normal morphology, expression of pluripotent markers, stable karyotype, potential of trilineage differentiation and absence of GDE expression, making them valuable tools for modeling GSDIII disease in vitro, studying pathological mechanisms and investigating potential treatments.


Assuntos
Sistema da Enzima Desramificadora do Glicogênio , Doença de Depósito de Glicogênio Tipo III , Células-Tronco Pluripotentes Induzidas , Humanos , Doença de Depósito de Glicogênio Tipo III/metabolismo , Doença de Depósito de Glicogênio Tipo III/patologia , Células-Tronco Pluripotentes Induzidas/metabolismo , Fígado/patologia , Músculos/metabolismo , Músculos/patologia
5.
Front Cell Dev Biol ; 11: 1163427, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37250895

RESUMO

Introduction: Glycogen storage disease type III (GSDIII) is a rare genetic disease caused by mutations in the AGL gene encoding the glycogen debranching enzyme (GDE). The deficiency of this enzyme, involved in cytosolic glycogen degradation, leads to pathological glycogen accumulation in liver, skeletal muscles and heart. Although the disease manifests with hypoglycemia and liver metabolism impairment, the progressive myopathy is the major disease burden in adult GSDIII patients, without any curative treatment currently available. Methods: Here, we combined the self-renewal and differentiation capabilities of human induced pluripotent stem cells (hiPSCs) with cutting edge CRISPR/Cas9 gene editing technology to establish a stable AGL knockout cell line and to explore glycogen metabolism in GSDIII. Results: Following skeletal muscle cells differentiation of the edited and control hiPSC lines, our study reports that the insertion of a frameshift mutation in AGL gene results in the loss of GDE expression and persistent glycogen accumulation under glucose starvation conditions. Phenotypically, we demonstrated that the edited skeletal muscle cells faithfully recapitulate the phenotype of differentiated skeletal muscle cells of hiPSCs derived from a GSDIII patient. We also demonstrated that treatment with recombinant AAV vectors expressing the human GDE cleared the accumulated glycogen. Discussion: This study describes the first skeletal muscle cell model of GSDIII derived from hiPSCs and establishes a platform to study the mechanisms that contribute to muscle impairments in GSDIII and to assess the therapeutic potential of pharmacological inducers of glycogen degradation or gene therapy approaches.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA