Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Acta Neuropathol Commun ; 6(1): 119, 2018 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-30404653

RESUMEN

Pompe disease is a metabolic myopathy that is caused by glycogen accumulation as a result of deficiency of the lysosomal enzyme acid alpha glucosidase (GAA). Previously, we showed that adult muscle stem cells termed satellite cells are present at normal levels in muscle from patients with Pompe disease, but that these are insufficiently activated to repair the severe muscle pathology. Here we characterized the muscle regenerative response during disease progression in a mouse model of Pompe disease and investigated the intrinsic capacity of Gaa-/- satellite cells to regenerate muscle damage. Gaa-/- mice showed progressive muscle pathology from 15 weeks of age as reflected by increased lysosomal size, decreased fiber diameter and reduced muscle wet weight. Only during the first 15 weeks of life but not thereafter, we detected a gradual increase in centrally nucleated fibers and proliferating satellite cells in Gaa-/- muscle, indicating a mild regenerative response. The levels of Pax7-positive satellite cells were increased in Gaa-/- mice at all ages, most likely as result of enhanced satellite cell activation in young Gaa-/- animals. Surprisingly, both young and old Gaa-/- mice regenerated experimentally-induced muscle injury efficiently as judged by rapid satellite cell activation and complete restoration of muscle histology. In response to serial injury, Gaa-/- mice also regenerated muscle efficiently and maintained the satellite cell pool. These findings suggest that, similar to human patients, Gaa-/- mice have insufficient satellite cell activation and muscle regeneration during disease progression. The initial endogenous satellite cell response in Gaa-/- mice may contribute to the delayed onset of muscle wasting compared to human patients. The rapid and efficient regeneration after experimental muscle injury suggest that Gaa-/- satellite cells are functional stem cells, opening avenues for developing muscle regenerative therapies for Pompe disease.


Asunto(s)
Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Músculo Esquelético/fisiopatología , Regeneración/genética , Células Satélite del Músculo Esquelético/fisiología , Factores de Edad , Animales , Compuestos de Bario/toxicidad , Cardiotoxinas/toxicidad , Cloruros/toxicidad , Modelos Animales de Enfermedad , Femenino , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/genética , Antígeno Ki-67/metabolismo , Laminina/metabolismo , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Músculo Esquelético/efectos de los fármacos , Factor de Transcripción PAX7/metabolismo , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo
2.
Sci Adv ; 4(9): eaar3938, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30258985

RESUMEN

The mechanistic target of rapamycin (mTOR) serine/threonine kinase, a critical regulator of cell proliferation, is frequently deregulated in human cancer. Although rapamycin inhibits the two canonical mTOR complexes, mTORC1 and mTORC2, it often shows minimal benefit as an anticancer drug. This is caused by rapamycin resistance of many different tumors, and we show that a third mTOR complex, mTORC3, contributes to this resistance. The ETS (E26 transformation-specific) transcription factor ETV7 interacts with mTOR in the cytoplasm and assembles mTORC3, which is independent of ETV7's transcriptional activity. This complex exhibits bimodal mTORC1/2 activity but is devoid of crucial mTORC1/2 components. Many human cancers activate mTORC3 at considerable frequency, and tumor cell lines that lose mTORC3 expression become rapamycin-sensitive. We show mTORC3's tumorigenicity in a rhabdomyosarcoma mouse model in which transgenic ETV7 expression accelerates tumor onset and promotes tumor penetrance. Discovery of mTORC3 represents an mTOR paradigm shift and identifies a novel target for anticancer drug development.


Asunto(s)
Neoplasias/metabolismo , Proteínas Proto-Oncogénicas c-ets/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Linfocitos B/efectos de los fármacos , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina/genética , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Proteínas Proto-Oncogénicas c-ets/genética , Proteína Asociada al mTOR Insensible a la Rapamicina/genética , Proteína Asociada al mTOR Insensible a la Rapamicina/metabolismo , Proteína Reguladora Asociada a mTOR/genética , Proteína Reguladora Asociada a mTOR/metabolismo , Transducción de Señal , Sirolimus/farmacología , Serina-Treonina Quinasas TOR/genética , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Leuk Res ; 39(8): 906-12, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26111797

RESUMEN

The MN1 (Meningioma 1) gene is overexpressed in certain subtypes of acute myeloid leukemia (AML) and high levels of MN1 expression in mouse bone marrow cells results in myeloid leukemia. We showed that compared with control bone marrow (BM) MN1 expression was increased (2-fold or more) in 29 out of 73 (40%) pediatric B-cell acute lymphoblastic leukemia (B-ALL) patient BM. Additional analysis of MN1 expression in sub-groups within our cohort carrying different chromosome translocations showed that carriers of the good prognostic marker t(12;21)(TEL-AML1) (n=27) expressed significantly more MN1 than both healthy controls (n=9) (P=0.02) and the group carrying the t(9;22)(BCR-ABL) (n=9) (P=0.001). In addition, AML1 expression was also upregulated in 31 out of 45 (68%) B-ALL patient BM compared with control and there was a significant correlation between MN1 and AML1 expression (r=0.3552, P=0.0167). Retroviral MN1 overexpression increased the colony forming activity of mouse Pro-B/Pre-B cells in vitro. Our results suggest that deregulated MN1 expression contributes to the pathogenesis of pediatric B-ALL. Further investigation into the clinical and biological significance of elevated MN1 expression in TEL-AML1(positive) leukemia might provide insight into additional molecular mechanisms contributing to B-ALL and may lead to improved treatment options for patients.


Asunto(s)
Linfocitos B/fisiología , Células de la Médula Ósea/fisiología , Células Madre Hematopoyéticas/fisiología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Proteínas Supresoras de Tumor/genética , Adolescente , Animales , Células de la Médula Ósea/metabolismo , Estudios de Casos y Controles , Células Cultivadas , Niño , Preescolar , Ensayo de Unidades Formadoras de Colonias , Femenino , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Humanos , Lactante , Masculino , Ratones , Ratones Endogámicos C57BL , Cultivo Primario de Células , Transactivadores , Adulto Joven
4.
EMBO Mol Med ; 5(5): 691-706, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23606558

RESUMEN

A recently proposed therapeutic approach for lysosomal storage disorders (LSDs) relies upon the ability of transcription factor EB (TFEB) to stimulate autophagy and induce lysosomal exocytosis leading to cellular clearance. This approach is particularly attractive in glycogen storage disease type II [a severe metabolic myopathy, Pompe disease (PD)] as the currently available therapy, replacement of the missing enzyme acid alpha-glucosidase, fails to reverse skeletal muscle pathology. PD, a paradigm for LSDs, is characterized by both lysosomal abnormality and dysfunctional autophagy. Here, we show that TFEB is a viable therapeutic target in PD: overexpression of TFEB in a new muscle cell culture system and in mouse models of the disease reduced glycogen load and lysosomal size, improved autophagosome processing, and alleviated excessive accumulation of autophagic vacuoles. Unexpectedly, the exocytosed vesicles were labelled with lysosomal and autophagosomal membrane markers, suggesting that TFEB induces exocytosis of autophagolysosomes. Furthermore, the effects of TFEB were almost abrogated in the setting of genetically suppressed autophagy, supporting the role of autophagy in TFEB-mediated cellular clearance.


Asunto(s)
Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/enzimología , Adenoviridae/genética , Animales , Autofagia , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/química , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Células Cultivadas , Modelos Animales de Enfermedad , Exocitosis , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Glucógeno/metabolismo , Enfermedad del Almacenamiento de Glucógeno Tipo II/patología , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Músculo Esquelético/citología , Músculo Esquelético/metabolismo , Músculo Esquelético/ultraestructura , alfa-Glucosidasas/deficiencia , alfa-Glucosidasas/genética , alfa-Glucosidasas/metabolismo
5.
Mol Ther ; 17(6): 964-71, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19293774

RESUMEN

In spite of the progress in the treatment of lysosomal storage diseases (LSDs), in some of these disorders the available therapies show limited efficacy and a need exists to identify novel therapeutic strategies. We studied the combination of enzyme replacement and enzyme enhancement by pharmacological chaperones in Pompe disease (PD), a metabolic myopathy caused by the deficiency of the lysosomal acid alpha-glucosidase. We showed that coincubation of Pompe fibroblasts with recombinant human alpha-glucosidase and the chaperone N-butyldeoxynojirimycin (NB-DNJ) resulted in more efficient correction of enzyme activity. The chaperone improved alpha-glucosidase delivery to lysosomes, enhanced enzyme maturation, and increased enzyme stability. Improved enzyme correction was also found in vivo in a mouse model of PD treated with coadministration of single infusions of recombinant human alpha-glucosidase and oral NB-DNJ. The enhancing effect of chaperones on recombinant enzymes was also observed in fibroblasts from another lysosomal disease, Fabry disease, treated with recombinant alpha-galactosidase A and the specific chaperone 1-deoxygalactonojirimycin (DGJ). These results have important clinical implications, as they demonstrate synergy between pharmacological chaperones and enzyme replacement. A synergistic effect of these treatments may result particularly useful in patients responding poorly to therapy and in tissues in which sufficient enzyme levels are difficult to obtain.


Asunto(s)
1-Desoxinojirimicina/análogos & derivados , Inhibidores Enzimáticos/uso terapéutico , Fibroblastos/efectos de los fármacos , Enfermedad del Almacenamiento de Glucógeno Tipo II/tratamiento farmacológico , 1-Desoxinojirimicina/farmacología , 1-Desoxinojirimicina/uso terapéutico , Animales , Transporte Biológico/efectos de los fármacos , Western Blotting , Línea Celular , Estabilidad de Medicamentos , Inhibidores Enzimáticos/farmacología , Fibroblastos/patología , Humanos , Lisosomas/metabolismo , Ratones , Microscopía Confocal , alfa-Galactosidasa/administración & dosificación , alfa-Galactosidasa/metabolismo , alfa-Galactosidasa/farmacología , alfa-Galactosidasa/uso terapéutico
6.
Pathogenetics ; 1(1): 6, 2008 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-19046416

RESUMEN

BACKGROUND: Pompe disease (PD) is a metabolic myopathy caused by alpha-glucosidase (GAA) deficiency and characterized by generalized glycogen storage. Heterogeneous GAA gene mutations result in wide phenotypic variability, ranging from the severe classic infantile presentation to the milder intermediate and late-onset forms. Enzyme replacement therapy (ERT) with recombinant human GAA (rhGAA), the only treatment available for PD, intriguingly shows variable efficacy in different PD patients. To investigate the mechanisms underlying the variable response to ERT, we studied cell morphology of PD fibroblasts, the distribution and trafficking of the cation-independent mannose-6-phosphate receptor (CI-MPR) that mediates rhGAA uptake, and rhGAA uptake itself. RESULTS: We observed abnormalities of cell morphology in PD cells. Electron microscopy analysis showed accumulation of multivesicular bodies and expansion of the Golgi apparatus, and immunolocalization and western blot analysis of LC3 showed activation of autophagy. Immunofluorescence analysis showed abnormal intracellular distribution of CI-MPR in PD fibroblasts, increased co-localization with LC3 and reduced availability of the receptor at the plasma membrane. The recycling of CI-MPR from the plasma membrane to the trans-Golgi network was also impaired. All these abnormalities were more prominent in severe and intermediate PD fibroblasts, correlating with disease severity. In severe and intermediate PD cells rhGAA uptake and processing were less efficient and correction of GAA activity was reduced. CONCLUSION: These results indicate a role for disrupted CI-MPR trafficking in the variable response to ERT in PD and have implications for ERT efficacy and optimization of treatment protocols.

7.
Curr Opin Neurol ; 20(2): 151-8, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17351484

RESUMEN

PURPOSE OF REVIEW: The use of gene therapy to correct or replace deficient genes has been a long-standing aspiration. RECENT FINDINGS: Recent findings from basic and applied research suggest that at last it may be possible to translate experimental procedures into effective patient therapies for genetic diseases. Therapies for neurodegenerative diseases potentially include, as their targets, both monogenic conditions (e.g. lysosomal storage disorders) and more genetically complex diseases (such as Alzheimer's and Parkinson's disorders). SUMMARY: The use of gene therapy to target the central nervous system presents specific technical and biological challenges. These may be overcome by using novel gene vector delivery strategies. Current research should illuminate the temporal window required to achieve a successful therapy. As greater knowledge is accumulated about gene therapy, correlations will be made between the level of gene expression from the therapeutic vector, the extent of correction after treatment, and the stage of disease progression when therapy is initiated.


Asunto(s)
Terapia Genética/métodos , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/terapia , Animales , Terapia Genética/tendencias , Humanos
8.
Hum Mol Genet ; 15(7): 1225-36, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16505002

RESUMEN

Mucopolysaccharidosis type II (MPSII; Hunter syndrome) is a lysosomal storage disorder caused by a deficiency in the enzyme iduronate 2-sulfatase (IDS). At present, the therapeutic approaches for MPSII are enzyme replacement therapy and bone marrow transplantation, although these therapies have some limitations. The availability of new AAV serotypes that display tissue-specific tropism and promote sustained expression of transgenes offers the possibility of AAV-mediated gene therapy for the systemic treatment of lysosomal diseases, including MPSII. We have characterized in detail the phenotype of IDS-deficient mice, a model of human MPSII. These mice display a progressive accumulation of glycosaminoglycans (GAGs) in many organs and excessive excretion of these compounds in their urine. Furthermore, they develop skeleton deformities, particularly of the craniofacial bones, and alopecia, they perform poorly in open-field tests and they have a severely compromised walking pattern. In addition, they present neuropathological defects. We have designed an efficient gene therapy approach for the treatment of these MPSII mice. AAV2/8TBG-IDS viral particles were administrated intravenously to adult MPSII mice. The plasma and tissue IDS activities were completely restored in all of the treated mice. This rescue of the enzymatic activity resulted in the full clearance of the accumulated GAGs in all of the tissues analyzed, the normalization of the GAG levels in the urine and the correction of the skeleton malformations. Overall, our findings suggest that this in vivo gene transfer approach has potential for the systemic treatment of patients with Hunter syndrome.


Asunto(s)
Dependovirus/genética , Modelos Animales de Enfermedad , Terapia Genética/métodos , Vectores Genéticos/uso terapéutico , Mucopolisacaridosis II/metabolismo , Animales , Dependovirus/metabolismo , Técnica del Anticuerpo Fluorescente , Vectores Genéticos/genética , Vectores Genéticos/metabolismo , Glicosaminoglicanos/metabolismo , Glicosaminoglicanos/orina , Humanos , Iduronato Sulfatasa/sangre , Iduronato Sulfatasa/genética , Iduronato Sulfatasa/metabolismo , Riñón/citología , Riñón/metabolismo , Hígado/citología , Hígado/metabolismo , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Actividad Motora , Mucopolisacaridosis II/terapia , Bazo/citología , Bazo/metabolismo , Proteínas de Unión a Tiroxina/genética , Proteínas de Unión a Tiroxina/metabolismo
9.
Mol Cell Biol ; 25(6): 2395-405, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15743832

RESUMEN

The human ETS family gene TEL2/ETV7 is highly homologous to TEL1/ETV6, a frequent target of chromosome translocations in human leukemia and specific solid tumors. Here we report that TEL2 augments the proliferation and survival of normal mouse B cells and dramatically accelerates lymphoma development in Emu-Myc transgenic mice. Nonetheless, inactivation of the p53 pathway was a hallmark of all TEL2/Emu-Myc lymphomas, indicating that TEL2 expression alone is insufficient to bypass this apoptotic checkpoint. Although TEL2 is infrequently up-regulated in human sporadic Burkitt's lymphoma, analysis of pediatric B-cell acute lymphocytic leukemia (B-ALL) samples showed increased coexpression of TEL2 and MYC and/or MYCN in over one-third of B-ALL patients. Therefore, TEL2 and MYC also appear to cooperate in provoking a cadre of human B-cell malignancies.


Asunto(s)
Linfoma de Burkitt/metabolismo , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Factores de Transcripción/metabolismo , Animales , Apoptosis , Linfocitos B/metabolismo , Linfoma de Burkitt/genética , Proliferación Celular , Niño , Proteínas de Unión al ADN/genética , Femenino , Humanos , Masculino , Ratones , Ratones Transgénicos , Mutación/genética , Proteínas Proto-Oncogénicas c-ets , Supresión Genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/genética , Regulación hacia Arriba/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...