Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 4 de 4
Filtrar
Mais filtros

Base de dados
Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 24(12): 3372-89, 2015 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-25749991

RESUMO

Globoid cell leukodystrophy (GLD) is a lysosomal storage disease caused by deficient activity of ß-galactocerebrosidase (GALC). The infantile forms manifest with rapid and progressive central and peripheral demyelination, which represent a major hurdle for any treatment approach. We demonstrate here that neonatal lentiviral vector-mediated intracerebral gene therapy (IC GT) or transplantation of GALC-overexpressing neural stem cells (NSC) synergize with bone marrow transplant (BMT) providing dramatic extension of lifespan and global clinical-pathological rescue in a relevant GLD murine model. We show that timely and long-lasting delivery of functional GALC in affected tissues ensured by the exclusive complementary mode of action of the treatments underlies the outstanding benefit. In particular, the contribution of neural stem cell transplantation and IC GT during the early asymptomatic stage of the disease is instrumental to enhance long-term advantage upon BMT. We clarify the input of central nervous system, peripheral nervous system and periphery to the disease, and the relative contribution of treatments to the final therapeutic outcome, with important implications for treatment strategies to be tried in human patients. This study gives proof-of-concept of efficacy, tolerability and clinical relevance of the combined gene/cell therapies proposed here, which may constitute a feasible and effective therapeutic opportunity for children affected by GLD.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Galactosilceramidase/genética , Terapia Genética , Leucodistrofia de Células Globoides/genética , Animais , Apoptose/genética , Axônios/metabolismo , Axônios/patologia , Transplante de Medula Óssea , Encéfalo/metabolismo , Diferenciação Celular , Sistema Nervoso Central/metabolismo , Sistema Nervoso Central/fisiopatologia , Modelos Animais de Doenças , Ativação Enzimática , Galactosilceramidase/metabolismo , Gliose/genética , Gliose/metabolismo , Gliose/patologia , Sobrevivência de Enxerto , Humanos , Leucodistrofia de Células Globoides/diagnóstico , Leucodistrofia de Células Globoides/metabolismo , Leucodistrofia de Células Globoides/mortalidade , Leucodistrofia de Células Globoides/terapia , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Bainha de Mielina/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Sistema Nervoso Periférico/metabolismo , Sistema Nervoso Periférico/fisiopatologia , Transplante de Células-Tronco
2.
Blood ; 116(11): 1857-66, 2010 Sep 16.
Artigo em Inglês | MEDLINE | ID: mdl-20511539

RESUMO

The balance between survival and death in many cell types is regulated by small changes in the intracellular content of bioactive sphingolipids. Enzymes that either produce or degrade these sphingolipids control this equilibrium. The findings here described indicate that the lysosomal galactocerebrosidase (GALC) enzyme, defective in globoid cell leukodystrophy, is involved in the maintenance of a functional hematopoietic stem/progenitor cell (HSPC) niche by contributing to the control of the intracellular content of key sphingolipids. Indeed, we show that both insufficient and supraphysiologic GALC activity-by inherited genetic deficiency or forced gene expression in patients' cells and in the disease model-induce alterations of the intracellular content of the bioactive GALC downstream products ceramide and sphingosine, and thus affect HSPC survival and function and the functionality of the stem cell niche. Therefore, GALC and, possibly, other enzymes for the maintenance of niche functionality and health tightly control the concentration of these sphingolipids within HSPCs.


Assuntos
Medula Óssea/enzimologia , Galactosilceramidase/metabolismo , Células-Tronco Hematopoéticas/enzimologia , Nicho de Células-Tronco/enzimologia , Animais , Apoptose/efeitos dos fármacos , Medula Óssea/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Citometria de Fluxo , Galactosilceramidase/deficiência , Galactosilceramidase/genética , Genótipo , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Imunofenotipagem , Marcação In Situ das Extremidades Cortadas , Fator de Crescimento Insulin-Like I/farmacologia , Leucodistrofia de Células Globoides/enzimologia , Leucodistrofia de Células Globoides/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Esfingolipídeos/metabolismo , Nicho de Células-Tronco/metabolismo , Transfecção , Células U937
3.
Mol Ther Methods Clin Dev ; 2: 15038, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26509184

RESUMO

Globoid cell leukodystrophy (GLD) is a demyelinating lysosomal storage disease due to the deficiency of the galactocerebrosidase (GALC) enzyme. The favorable outcome of hematopoietic stem and progenitor cell (HSPC)-based approaches in GLD and other similar diseases suggests HSPC gene therapy as a promising therapeutic option for patients. The path to clinical development of this strategy was hampered by a selective toxicity of the overexpressed GALC in the HSPC compartment. Here, we presented the optimization of a lentiviral vector (LV) in which miR-126 regulation was coupled to codon optimization of the human GALC cDNA to obtain a selective and enhanced enzymatic activity only upon transduced HSPCs differentiation. The safety of human GALC overexpression driven by this LV was extensively demonstrated in vitro and in vivo on human HSPCs from healthy donors. No perturbation in the content of proapoptotic sphingolipids, gene expression profile, and capability of engraftment and mutlilineage differentiation in chimeric mice was observed. The therapeutic potential of this LV was then assessed in a severe GLD murine model that benefited from transplantation of corrected HSPCs with longer survival and ameliorated phenotype as compared to untreated siblings. This construct has thus been selected as a candidate for clinical translation.

4.
Sci Transl Med ; 2(58): 58ra84, 2010 Nov 17.
Artigo em Inglês | MEDLINE | ID: mdl-21084719

RESUMO

Globoid cell leukodystrophy (GLD; also known as Krabbe disease) is an invariably fatal lysosomal storage disorder caused by mutations in the galactocerebrosidase (GALC) gene. Hematopoietic stem cell (HSC)-based gene therapy is being explored for GLD; however, we found that forced GALC expression was toxic to HSCs and early progenitors, highlighting the need for improved regulation of vector expression. We used a genetic reporter strategy based on lentiviral vectors to detect microRNA activity in hematopoietic cells at single-cell resolution. We report that miR-126 and miR-130a were expressed in HSCs and early progenitors from both mice and humans, but not in differentiated progeny. Moreover, repopulating HSCs could be purified solely on the basis of miRNA expression, providing a new method relevant for human HSC isolation. By incorporating miR-126 target sequences into a GALC-expressing vector, we suppressed GALC expression in HSCs while maintaining robust expression in mature hematopoietic cells. This approach protected HSCs from GALC toxicity and allowed successful treatment of a mouse GLD model, providing a rationale to explore HSC-based gene therapy for GLD.


Assuntos
Terapia Genética , Células-Tronco Hematopoéticas/metabolismo , Leucodistrofia de Células Globoides/genética , Leucodistrofia de Células Globoides/terapia , MicroRNAs/metabolismo , Animais , Diferenciação Celular , Separação Celular , Citoproteção , Galactosilceramidase/metabolismo , Regulação da Expressão Gênica , Genes Transgênicos Suicidas , Células-Tronco Hematopoéticas/citologia , Humanos , Camundongos , Camundongos SCID , MicroRNAs/genética , Especificidade de Órgãos/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA