Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Más filtros




Base de datos
Intervalo de año de publicación
1.
Stem Cell Reports ; 9(2): 409-418, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28757166

RESUMEN

Dyskeratosis congenita (DC) is a bone marrow failure syndrome associated with telomere dysfunction. The progression and molecular determinants of hematopoietic failure in DC remain poorly understood. Here, we use the directed differentiation of human embryonic stem cells harboring clinically relevant mutations in telomerase to understand the consequences of DC-associated mutations on the primitive and definitive hematopoietic programs. Interestingly, telomere shortening does not broadly impair hematopoiesis, as primitive hematopoiesis is not impaired in DC cells. In contrast, while phenotypic definitive hemogenic endothelium is specified, the endothelial-to-hematopoietic transition is impaired in cells with shortened telomeres. This failure is caused by DNA damage accrual and is mediated by p53 stabilization. These observations indicate that detrimental effects of telomere shortening in the hematopoietic system are specific to the definitive hematopoietic lineages. This work illustrates how telomere dysfunction impairs hematopoietic development and creates a robust platform for therapeutic discovery for treatment of DC patients.


Asunto(s)
Disqueratosis Congénita/sangre , Disqueratosis Congénita/genética , Hematopoyesis/genética , Proteína p53 Supresora de Tumor/genética , Anemia Aplásica/sangre , Anemia Aplásica/etiología , Anemia Aplásica/patología , Biomarcadores , Médula Ósea/patología , Enfermedades de la Médula Ósea/sangre , Enfermedades de la Médula Ósea/etiología , Enfermedades de la Médula Ósea/patología , Trastornos de Fallo de la Médula Ósea , Diferenciación Celular/genética , Daño del ADN , Análisis Mutacional de ADN , Disqueratosis Congénita/patología , Células Madre Embrionarias/metabolismo , Técnicas de Inactivación de Genes , Marcación de Gen , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Hemoglobinuria Paroxística/sangre , Hemoglobinuria Paroxística/etiología , Hemoglobinuria Paroxística/patología , Histonas/metabolismo , Humanos , Inmunofenotipificación , Modelos Biológicos , Mutación , Fenotipo , Telómero , Homeostasis del Telómero/genética , Proteína p53 Supresora de Tumor/metabolismo
2.
J Exp Clin Cancer Res ; 32: 31, 2013 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-23701745

RESUMEN

Anticancer activities of cinnamic acid derivatives include induction of apoptosis by irreversible DNA damage leading to cell death. The present work aimed to compare the cytotoxic and genotoxic potential of cinnamic acid in human melanoma cell line (HT-144) and human melanocyte cell line derived from blue nevus (NGM). Viability assay showed that the IC50 for HT-144 cells was 2.4 mM, while NGM cells were more resistant to the treatment. The growth inhibition was probably associated with DNA damage leading to DNA synthesis inhibition, as shown by BrdU incorporation assay, induction of nuclear aberrations and then apoptosis. The frequency of cell death caused by cinnamic acid was higher in HT-144 cells. Activated-caspase 3 staining showed apoptosis after 24 hours of treatment with cinnamic acid 3.2 mM in HT-144 cells, but not in NGM. We observed microtubules disorganization after cinnamic acid exposure, but this event and cell death seem to be independent according to M30 and tubulin labeling. The frequency of micronucleated HT-144 cells was higher after treatment with cinnamic acid (0.4 and 3.2 mM) when compared to the controls. Cinnamic acid 3.2 mM also increased the frequency of micronucleated NGM cells indicating genotoxic activity of the compound, but the effects were milder. Binucleation and multinucleation counting showed similar results. We conclude that cinnamic acid has effective antiproliferative activity against melanoma cells. However, the increased frequency of micronucleation in NGM cells warrants the possibility of genotoxicity and needs further investigation.


Asunto(s)
Apoptosis/efectos de los fármacos , Cinamatos/farmacología , Melanoma/metabolismo , Caspasa 9/metabolismo , Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Aberraciones Cromosómicas/efectos de los fármacos , Cinamatos/toxicidad , Citoesqueleto/metabolismo , Humanos , Microtúbulos/metabolismo , Fosforilación/efectos de los fármacos , Tubulina (Proteína)/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
3.
Cancer Cell Int ; 13(1): 38, 2013 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-23631593

RESUMEN

BACKGROUND: Lung cancer often exhibits molecular changes, such as the overexpression of the ErbB1 gene. ErbB1 encodes epidermal growth factor receptor (EGFR), a tyrosine kinase receptor, involved mainly in cell proliferation and survival. EGFR overexpression has been associated with more aggressive disease, poor prognosis, low survival rate and low response to therapy. ErbB1 amplification and mutation are associated with tumor development and are implicated in ineffective treatment. The aim of the present study was to investigate whether the ErbB1 copy number affects EGFR expression, cell proliferation or cell migration by comparing two different cell lines. METHODS: The copies of ErbB1 gene was evaluated by FISH. Immunofluorescence and Western blotting were performed to determine location and expression of proteins mentioned in the present study. Proliferation was studied by flow cytometry and cell migration by wound healing assay and time lapse. RESULTS: We investigated the activation and function of EGFR in the A549 and HK2 lung cancer cell lines, which contain 3 and 6 copies of ErbB1, respectively. The expression of EGFR was lower in the HK2 cell line. EGFR was activated after stimulation with EGF in both cell lines, but this activation did not promote differences in cellular proliferation when compared to control cells. Inhibiting EGFR with AG1478 did not modify cellular proliferation, confirming previous data. However, we observed morphological alterations, changes in microfilament organization and increased cell migration upon EGF stimulation. However, these effects did not seem to be consequence of an epithelial-mesenchymal transition. CONCLUSION: EGFR expression did not appear to be associated to the ErbB1 gene copy number, and neither of these aspects appeared to affect cell proliferation. However, EGFR activation by EGF resulted in cell migration stimulation in both cell lines.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA