Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Gene Ther ; 29(9): 520-535, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35105949

RESUMO

Duchenne muscular dystrophy (DMD) is a muscle wasting disorder caused by mutations in the gene encoding dystrophin. Gene therapy using micro-dystrophin (MD) transgenes and recombinant adeno-associated virus (rAAV) vectors hold great promise. To overcome the limited packaging capacity of rAAV vectors, most MD do not include dystrophin carboxy-terminal (CT) domain. Yet, the CT domain is known to recruit α1- and ß1-syntrophins and α-dystrobrevin, a part of the dystrophin-associated protein complex (DAPC), which is a signaling and structural mediator of muscle cells. In this study, we explored the impact of inclusion of the dystrophin CT domain on ΔR4-23/ΔCT MD (MD1), in DMDmdx rats, which allows for relevant evaluations at muscular and cardiac levels. We showed by LC-MS/MS that MD1 expression is sufficient to restore the interactions at a physiological level of most DAPC partners in skeletal and cardiac muscles, and that inclusion of the CT domain increases the recruitment of some DAPC partners at supra-physiological levels. In parallel, we demonstrated that inclusion of the CT domain does not improve MD1 therapeutic efficacy on DMD muscle and cardiac pathologies. Our work highlights new evidences of the therapeutic potential of MD1 and strengthens the relevance of this candidate for gene therapy of DMD.


Assuntos
Distrofina , Distrofia Muscular de Duchenne , Animais , Cromatografia Líquida , Distrofina/genética , Distrofina/metabolismo , Complexo de Proteínas Associadas Distrofina/metabolismo , Terapia Genética , Músculo Esquelético/metabolismo , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Ratos , Espectrometria de Massas em Tandem
2.
Hum Gene Ther ; 32(19-20): 1138-1146, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-33765840

RESUMO

Duchenne muscular dystrophy (DMD) is an X-linked recessive disease that affects 1:5,000 live male births and is characterized by muscle wasting. By the age of 13 years, affected individuals are often wheelchair bound and suffer from respiratory and cardiac failure, which results in premature death. Although the administration of corticosteroids and ventilation can relieve the symptoms and extend the patients' lifespan, currently no cure exists for DMD. Among the different approaches under preclinical and clinical testing, gene therapy, using adeno-associated viral (AAV) vectors, is one of the most promising. In this study, we delivered intravenously AAV9 vectors expressing the microdystrophin MD1 (ΔR4-R23/ΔCT) under control of the synthetic muscle-specific promoter Spc5-12 and assessed the effect of adding a cardiac-specific cis-regulatory module (designated as CS-CRM4) on its expression profile in skeletal and cardiac muscles. Results show that Spc5-12 promoter, in combination with an AAV serotype that has high tropism for the heart, drives high MD1 expression levels in cardiac muscle in mdx mice. The additional regulatory element CS-CRM4 can further improve MD1 expression in cardiac muscles, but its effect is dose dependent and enhancement becomes evident only at lower vector doses.


Assuntos
Distrofina , Distrofia Muscular de Duchenne , Animais , Dependovirus/genética , Distrofina/genética , Vetores Genéticos/genética , Humanos , Masculino , Camundongos , Camundongos Endogâmicos mdx , Músculo Esquelético , Distrofia Muscular de Duchenne/genética , Distrofia Muscular de Duchenne/terapia , Miocárdio
3.
Front Immunol ; 11: 1398, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32733464

RESUMO

The bioavailability of the major pro-inflammatory cytokines IL-1α and IL-1ß is tightly controlled by transcription and post-translational processing to prevent hyperinflammation. The role of mRNA decay in maintenance of physiological IL-1 amounts remained unknown. Here we show that the down-regulation of Il1a and Il1b mRNA by the mRNA-destabilizing protein TTP (gene Zfp36) is required for immune homeostasis. The TTP deficiency syndrome, a multi organ inflammation in TTP-/- mice, was significantly ameliorated upon deletion of the IL-1 receptor. Il1a and Il1b played non-redundant roles in triggering the pathological IL-1 signaling in TTP-/- mice. Accordingly, tissues from TTP-/- animals contained increased amounts of Il1b mRNA. Unexpectedly, TTP destabilized Il1b mRNA in cell type-specific ways as evident from RNA-Seq and mRNA stability assays. These results demonstrate that TTP-driven mRNA destabilization depends on the cellular context. Moreover, such context-defined mRNA decay is essential for keeping steady state IL-1 levels in the physiological range.


Assuntos
Regulação da Expressão Gênica , Homeostase , Imunidade/genética , Interleucina-1/genética , Tristetraprolina/metabolismo , Animais , Modelos Animais de Doenças , Ensaio de Imunoadsorção Enzimática , Perfilação da Expressão Gênica , Inflamação/diagnóstico , Inflamação/etiologia , Inflamação/metabolismo , Interleucina-1/metabolismo , Interleucina-1alfa/genética , Interleucina-1beta/genética , Camundongos , Camundongos Knockout , Estabilidade de RNA , Reação em Cadeia da Polimerase em Tempo Real , Índice de Gravidade de Doença
4.
Hum Gene Ther ; 28(3): 242-254, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27889981

RESUMO

This study describes the initial testing of a novel strategy for neutralization of lentiviruses using the fundamental biology of enveloped viruses' assembly and budding. In the field of gene therapy, viral vector surface proteins have been manipulated in order to redirect host cell specificity by alteration of pseudo-types. This study tested whether known viral pseudo-typing proteins or surface proteins known to be recruited to the human immunodeficiency virus (HIV) envelope could be engineered to carry neutralizing epitopes from another microorganism onto the lentiviral surface. The results identify ICAM1 as a novel vehicle for lentiviral pseudo-typing. Importantly, the study shows that in a model lentiviral system, ICAM1 can be engineered in chimeric form to result in expression of a fragment of the tetanus toxoid on the viral membrane and that these viruses can then be neutralized by human serum antibodies protective against tetanus. This raises the possibility of delivering chimeric antigens as a gene therapy in HIV-infected patients.


Assuntos
Anticorpos Neutralizantes/farmacologia , Membrana Celular/imunologia , Infecções por HIV/terapia , Molécula 1 de Adesão Intercelular/imunologia , Lentivirus/imunologia , Tétano/imunologia , Epitopos/imunologia , Células HEK293 , Proteína gp120 do Envelope de HIV/imunologia , Infecções por HIV/imunologia , Infecções por HIV/virologia , HIV-1/isolamento & purificação , Humanos , Testes de Neutralização
5.
Hum Mol Genet ; 25(16): 3555-3563, 2016 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-27378686

RESUMO

In preclinical models for Duchenne muscular dystrophy, dystrophin restoration during adeno-associated virus (AAV)-U7-mediated exon-skipping therapy was shown to decrease drastically after six months in treated muscles. This decline in efficacy is strongly correlated with the loss of the therapeutic AAV genomes, probably due to alterations of the dystrophic myofiber membranes. To improve the membrane integrity of the dystrophic myofibers at the time of AAV-U7 injection, mdx muscles were pre-treated with a single dose of the peptide-phosphorodiamidate morpholino (PPMO) antisense oligonucleotides that induced temporary dystrophin expression at the sarcolemma. The PPMO pre-treatment allowed efficient maintenance of AAV genomes in mdx muscles and enhanced the AAV-U7 therapy effect with a ten-fold increase of the protein level after 6 months. PPMO pre-treatment was also beneficial to AAV-mediated gene therapy with transfer of micro-dystrophin cDNA into muscles. Therefore, avoiding vector genome loss after AAV injection by PPMO pre-treatment would allow efficient long-term restoration of dystrophin and the use of lower and thus safer vector doses for Duchenne patients.


Assuntos
Distrofina/genética , Terapia Genética , Morfolinos/administração & dosagem , Distrofia Muscular Animal/terapia , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/administração & dosagem , Animais , Dependovirus/genética , Éxons/genética , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Humanos , Camundongos Endogâmicos mdx , Músculo Esquelético/efeitos dos fármacos , Músculo Esquelético/patologia , Distrofia Muscular Animal/genética , Distrofia Muscular de Duchenne/genética , Sarcolema/efeitos dos fármacos , Sarcolema/patologia
6.
Curr Gene Ther ; 15(4): 395-415, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26159373

RESUMO

Duchenne muscular dystrophy (DMD), an X-linked inherited musclewasting disease primarily affecting young boys with prevalence of between1:3,500- 1:5,000, is a rare genetic disease caused by defects in the gene for dystrophin. Dystrophin protein is critical to the stability of myofibers in skeletal and cardiac muscle. There is currently no cure available to ameliorate DMD and/or its patho-physiology. A number of therapeutic strategies including molecular-based therapeutics that replace or correct the missing or nonfunctional dystrophin protein have been devised to correct the patho-physiological consequences induced by dystrophin absence. We will review the current in vivo experimentation status (including preclinical models and clinical trials) for two of these approaches, namely: 1) Adeno-associated virus (AAV) mediated (micro) dystrophin gene augmentation/ supplementation and 2) Antisense oligonucleotide (AON)-mediated exon skipping strategies.


Assuntos
Dependovirus/genética , Distrofina/genética , Terapia Genética/métodos , Distrofia Muscular de Duchenne/etiologia , Distrofia Muscular de Duchenne/terapia , Oligonucleotídeos Antissenso/farmacologia , Animais , Ensaios Clínicos como Assunto , Dependovirus/imunologia , Modelos Animais de Doenças , Distrofina/deficiência , Éxons , Técnicas de Transferência de Genes , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Humanos , Distrofia Muscular de Duchenne/genética , Oligonucleotídeos Antissenso/genética , Oxidiazóis/farmacologia , Edição de RNA
7.
Sci Rep ; 5: 11696, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-26114395

RESUMO

Spinal muscular atrophy (SMA) is a neuromuscular disease caused by mutations in Survival Motor Neuron 1 (SMN1), leading to degeneration of alpha motor neurons (MNs) but also affecting other cell types. Induced pluripotent stem cell (iPSC)-derived human MN models from severe SMA patients have shown relevant phenotypes. We have produced and fully characterized iPSCs from members of a discordant consanguineous family with chronic SMA. We differentiated the iPSC clones into ISL-1+/ChAT+ MNs and performed a comparative study during the differentiation process, observing significant differences in neurite length and number between family members. Analyses of samples from wild-type, severe SMA type I and the type IIIa/IV family showed a progressive decay in SMN protein levels during iPSC-MN differentiation, recapitulating previous observations in developmental studies. PLS3 underwent parallel reductions at both the transcriptional and translational levels. The underlying, progressive developmental decay in SMN and PLS3 levels may lead to the increased vulnerability of MNs in SMA disease. Measurements of SMN and PLS3 transcript and protein levels in iPSC-derived MNs show limited value as SMA biomarkers.


Assuntos
Diferenciação Celular , Células-Tronco Pluripotentes Induzidas/citologia , Glicoproteínas de Membrana/metabolismo , Proteínas dos Microfilamentos/metabolismo , Neurônios Motores/metabolismo , Neurônios Motores/patologia , Proteína 1 de Sobrevivência do Neurônio Motor/metabolismo , Animais , Biomarcadores/metabolismo , Sobrevivência Celular , Células Clonais , Técnicas de Cocultura , Feminino , Humanos , Masculino , Camundongos , Fibras Musculares Esqueléticas/citologia , Neuritos/metabolismo , Linhagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA