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

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Ther ; 28(6): 1442-1454, 2020 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-32278382

RESUMEN

Our previous study delivered zinc finger nucleases to treat mice with mucopolysaccharidosis type I (MPS I), resulting in a phase I/II clinical trial (ClinicalTrials.gov: NCT02702115). However, in the clinical trial, the efficacy needs to be improved due to the low transgene expression level. To this end, we designed a proprietary system (PS) gene editing approach with CRISPR to insert a promoterless α-l-iduronidase (IDUA) cDNA sequence into the albumin locus of hepatocytes. In this study, adeno-associated virus 8 (AAV8) vectors delivering the PS gene editing system were injected into neonatal and adult MPS I mice. IDUA enzyme activity in the brain significantly increased, while storage levels were normalized. Neurobehavioral tests showed that treated mice had better memory and learning ability. Also, histological analysis showed efficacy reflected by the absence of foam cells in the liver and vacuolation in neuronal cells. No vector-associated toxicity or increased tumorigenesis risk was observed. Moreover, no off-target effects were detected through the unbiased genome-wide unbiased identification of double-stranded breaks enabled by sequencing (GUIDE-seq) analysis. In summary, these results showed the safety and efficacy of the PS in treating MPS I and paved the way for clinical studies. Additionally, as a therapeutic platform, the PS has the potential to treat other lysosomal diseases.


Asunto(s)
Edición Génica/métodos , Expresión Génica , Terapia Genética , Iduronidasa/genética , Mucopolisacaridosis I/genética , Mucopolisacaridosis I/terapia , Transgenes , Animales , Encéfalo/metabolismo , Sistemas CRISPR-Cas , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Dependovirus/genética , Modelos Animales de Enfermedad , Activación Enzimática , Dosificación de Gen , Orden Génico , Técnicas de Transferencia de Gen , Terapia Genética/efectos adversos , Terapia Genética/métodos , Vectores Genéticos/genética , Humanos , Hígado/metabolismo , Hígado/patología , Ratones , Mucopolisacaridosis I/metabolismo , ARN Guía de Kinetoplastida , Resultado del Tratamiento
2.
Gene Ther ; 27(5): 226-236, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-31896760

RESUMEN

The GM2-gangliosidoses are neurological diseases causing premature death, thus developing effective treatment protocols is urgent. GM2-gangliosidoses result from deficiency of a lysosomal enzyme ß-hexosaminidase (Hex) and subsequent accumulation of GM2 gangliosides. Genetic changes in HEXA, encoding the Hex α subunit, or HEXB, encoding the Hex ß subunit, causes Tay-Sachs disease and Sandhoff disease, respectively. Previous studies have showed that a modified human Hex µ subunit (HEXM) can treat both Tay-Sachs and Sandhoff diseases by forming a homodimer to degrade GM2 gangliosides. To this end, we applied this HEXM subunit in our PS813 gene editing system to treat neonatal Sandhoff mice. Through AAV delivery of the CRISPR system, a promoterless HEXM cDNA will be integrated into the albumin safe harbor locus, and lysosomal enzyme will be expressed and secreted from edited hepatocytes. 4 months after the i.v. of AAV vectors, plasma MUGS and MUG activities reached up to 144- and 17-fold of wild-type levels (n = 10, p < 0.0001), respectively. More importantly, MUGS and MUG activities in the brain also increased significantly compared with untreated Sandhoff mice (p < 0.001). Further, HPLC-MS/MS analysis showed that GM2 gangliosides in multiple tissues, except the brain, of treated mice were reduced to normal levels. Rotarod analysis showed that coordination and motor memory of treated mice were improved (p < 0.05). Histological analysis of H&E stained tissues showed reduced cellular vacuolation in the brain and liver of treated Sandhoff mice. These results demonstrate the potential of developing a treatment of in vivo genome editing for Tay-Sachs and Sandhoff patients.


Asunto(s)
Enfermedad de Sandhoff , Enfermedad de Tay-Sachs , Animales , Modelos Animales de Enfermedad , Edición Génica , Humanos , Ratones , Enfermedad de Sandhoff/genética , Enfermedad de Sandhoff/terapia , Espectrometría de Masas en Tándem , Enfermedad de Tay-Sachs/genética , Enfermedad de Tay-Sachs/terapia , beta-N-Acetilhexosaminidasas/genética
3.
Carcinogenesis ; 33(11): 2076-83, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22847180

RESUMEN

Chondrosarcoma is a form of malignant skeletal tumor of cartilaginous origin. The non-malignant form of the disease is termed chondroma. Correctly distinguishing between the two forms is essential for making therapeutic decisions. However, due to their similar histological appearances and the lack of a reliable diagnostic marker, it is often difficult to distinguish benign tumors from low-grade chondrosarcoma. Therefore, it is necessary to search for a potential marker that has diagnostic and prognostic values in chondrosarcoma. In this study, we demonstrated by immunohistochemistry that elevated leukemia/lymphoma-related factor (LRF) expression was associated with increased malignancy in human chondrosarcoma tissue microarrays. Moreover, siRNA depletion of LRF drastically reduced proliferation of chondrosarcoma cell lines and effectively induced senescence in these cells. This could be attributed to the observation that LRF-depleted cells were arrested at the G(1) phase, and had increased p53 and p21 expression. Moreover, LRF depletion not only drastically reduces the cellular migration and invasion potentials of chondrosarcoma cells but also sensitized these cells to the apoptosis-inducing chemotherapeutic agent doxorubicin. We conclude that LRF is a survival factor in chondrosarcomas and its expression correlates with tumor malignancy and chemoresistance. Our data implicate the potential role of LRF as both a diagnostic marker and therapeutic target for chondrosarcomas.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Óseas/patología , Condrosarcoma/patología , Proteínas de Unión al ADN/metabolismo , Doxorrubicina/farmacología , Resistencia a Antineoplásicos , Oncogenes , Factores de Transcripción/metabolismo , Antibióticos Antineoplásicos/farmacología , Western Blotting , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/metabolismo , Adhesión Celular/efectos de los fármacos , Ciclo Celular/efectos de los fármacos , Movimiento Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Condrosarcoma/tratamiento farmacológico , Condrosarcoma/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Humanos , Técnicas para Inmunoenzimas , Clasificación del Tumor , Pronóstico , ARN Interferente Pequeño/genética , Análisis de Matrices Tisulares , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/metabolismo
4.
Sci Rep ; 6: 39059, 2016 12 14.
Artículo en Inglés | MEDLINE | ID: mdl-27966608

RESUMEN

Osteosarcoma is the most common primary bone tumor, with metastatic disease responsible for most treatment failure and patient death. A forward genetic screen utilizing Sleeping Beauty mutagenesis in mice previously identified potential genetic drivers of osteosarcoma metastasis, including Slit-Robo GTPase-Activating Protein 2 (Srgap2). This study evaluates the potential role of SRGAP2 in metastases-associated properties of osteosarcoma cell lines through Srgap2 knockout via the CRISPR/Cas9 nuclease system and conditional overexpression in the murine osteosarcoma cell lines K12 and K7M2. Proliferation, migration, and anchorage independent growth were evaluated. RNA sequencing and immunohistochemistry of human osteosarcoma tissue samples were used to further evaluate the potential role of the Slit-Robo pathway in osteosarcoma. The effects of Srgap2 expression modulation in the murine OS cell lines support the hypothesis that SRGAP2 may have a role as a suppressor of metastases in osteosarcoma. Additionally, SRGAP2 and other genes in the Slit-Robo pathway have altered transcript levels in a subset of mouse and human osteosarcoma, and SRGAP2 protein expression is reduced or absent in a subset of primary tumor samples. SRGAP2 and other axon guidance proteins likely play a role in osteosarcoma metastasis, with loss of SRGAP2 potentially contributing to a more aggressive phenotype.


Asunto(s)
Neoplasias Óseas/metabolismo , Proteínas Activadoras de GTPasa/genética , Proteínas Activadoras de GTPasa/metabolismo , Genes Supresores de Tumor , Osteosarcoma/metabolismo , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Técnicas de Inactivación de Genes , Pruebas Genéticas , Humanos , Ratones , Clasificación del Tumor , Metástasis de la Neoplasia , Osteosarcoma/genética , Osteosarcoma/patología , Análisis de Secuencia de ARN
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA