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

Base de dados
Ano de publicação
Tipo de documento
Intervalo de ano de publicação
1.
PLoS One ; 11(5): e0154309, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27148974

RESUMO

Pluripotent stem cells of the early embryo, and germ line cells, are essential to ensure uncompromised development to adulthood as well as species propagation, respectively. Recently, the transcription factor hypoxia inducible factor 1 alpha (Hif1α) has been shown to have important roles in embryonic stem cells; in particular, regulation of conversion to glycolytic metabolism and, as we have shown, maintenance of functional levels of telomerase. In the present study, we sought to assess whether Hif1α was also expressed in the primitive cells of the murine embryo. We observed expression of Hif1α in pre-implantation embryos, specifically the 2-cell stage, morula, and blastocyst. Robust Hif1α expression was also observed in male and female primordial germ cells. We subsequently assessed whether Hif1α was expressed in adult male and female germ cells. In the testis, Hif1α was robustly expressed in spermatogonial cells, in both juvenile (6-week old) and adult (3-month old) males. In the ovaries, Hif1α was expressed in mature oocytes from adult females, as assessed both in situ and in individual oocytes flushed from super-ovulated females. Analysis of Hif1α transcript levels indicates a mechanism of regulation during early development that involves stockpiling of Hif1α protein in mature oocytes, presumably to provide protection from hypoxic stress until the gene is re-activated at the blastocyst stage. Together, these observations show that Hif1α is expressed throughout the life-cycle, including both the male and female germ line, and point to an important role for Hif1α in early progenitor cells.


Assuntos
Células Germinativas/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Animais , Western Blotting , Feminino , Imunofluorescência , Subunidade alfa do Fator 1 Induzível por Hipóxia/biossíntese , Masculino , Camundongos/embriologia , Ovário/embriologia , Ovário/crescimento & desenvolvimento , Ovário/metabolismo , Óvulo/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espermatozoides/metabolismo , Testículo/embriologia , Testículo/crescimento & desenvolvimento , Testículo/metabolismo
2.
Oncotarget ; 7(15): 20869-89, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26989079

RESUMO

Resident mesenchymal stem cells (MSCs) promote cancer progression. However, pathways and mechanisms involved in recruiting MSCs into breast tumors remain largely undefined. Here we show that geminin-dependent acetylation releases HMGB1 from the chromatin to the cytoplasm and extracellular space. Extracellular acetylated HMGB1 (Ac-HMGB1) promotes geminin overexpressing (GemOE) cells survival by binding to RAGE and activating NF-κB signaling. Extracellular Ac-HMGB1 also triggers expression and activation of RAGE in the non-expressing MSCs. RAGE activation induces expression of CXCR4 in MSCs and directional migration towards SDF1 (aka CXCL12)-expressing GemOE cells in vitro and in vivo. These effects augmented by the necrotic and hypoxic environment in GemOE tumors, especially within their cores. Reciprocal interactions between newly recruited MSCs and GemOE tumor cells elevate tumor-initiating (TIC), basal and epithelial-to-mesenchymal transition (EMT) traits and enhance aggressiveness in vitro and in vivo in GemOE tumor cells. Indeed, faster, larger and more aggressive tumors develop when GemOE cells are co-injected with MSCs in orthotopic breast tumor model. Concurrently, inhibiting c-Abl (and thus geminin function), RAGE or CXCR4 prevented MSCs recruitment to GemOE cells in vitro and in vivo, and decreased the TIC, basal and EMT phenotypes in these tumor cells. Accordingly, we propose that GemOE tumor cells present within tumor cores represent metastatic precursors, and suppressing the GemOE→HMGB1/RAGE→SDF1/CXCR4 signaling circuit could be a valid target for therapies to inhibit GemOE tumors and their metastases.


Assuntos
Mama/patologia , Carcinoma Ductal de Mama/secundário , Transformação Celular Neoplásica/patologia , Geminina/metabolismo , Células-Tronco Mesenquimais/patologia , Neoplasias de Mama Triplo Negativas/patologia , Animais , Apoptose , Biomarcadores Tumorais/metabolismo , Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/metabolismo , Quimiocina CXCL12/metabolismo , Transição Epitelial-Mesenquimal , Feminino , Proteína HMGB1/metabolismo , Humanos , Metástase Linfática , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Nus , NF-kappa B/metabolismo , Invasividade Neoplásica , Prognóstico , Receptores CXCR4/metabolismo , Transdução de Sinais , Taxa de Sobrevida , Neoplasias de Mama Triplo Negativas/metabolismo , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Front Biosci (Landmark Ed) ; 21(2): 303-15, 2016 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-26709774

RESUMO

The cellular response to a hypoxic environment is regulated by hypoxia inducible factors. Hypoxia inducible factor 1 alpha (Hif1alpha) in particular, is tightly regulated by the hypoxic environment in most cells, and plays an important role in regulating the stress response of cells to hypoxia. Interestingly, substantial observations are now emerging that point to an important role for Hif1alpha in stem cells, including embryonic stem cells, neuronal stem cells and hematopoietic stem cells. Notably, Hif1alpha has been shown to enhance self renewal of stem cells, mediate a shift to glycolytic metabolism, and promote telomerase expression.


Assuntos
Hipóxia Celular , Células-Tronco/citologia , Telomerase/metabolismo , Senescência Celular , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Células-Tronco/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA