Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 5 de 5
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Genes Dev ; 24(24): 2800-11, 2010 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-21159820

RESUMEN

Organization into polarized three-dimensional structures defines whether epithelial cells are normal or malignant. In a model of morphogenesis, we show that inhibiting key signaling pathways in human breast cancer cells leads to "phenotypic reversion" of the malignant cells. Using architecture as an endpoint, we report that, in all cases, signaling through Raf/MEK/ERK disrupted tissue polarity via matrix metalloproteinase9 (MMP9) activity. Induction of Raf or activation of an engineered, functionally inducible MMP9 in nonmalignant cells led to loss of tissue polarity, and reinitiated proliferation. Conversely, inhibition of Raf or MMP9 with small molecule inhibitors or shRNAs restored the ability of cancer cells to form polarized quiescent structures. Silencing MMP9 expression also reduced tumor growth dramatically in a murine xenograft model. LC-MS/MS analysis comparing conditioned medium from nonmalignant cells with or without active MMP9 revealed laminin 111 (LM1) as an important target of MMP9. LM1 has been implicated in acinar morphogenesis; thus, its degradation by MMP9 provides a mechanism for loss of tissue polarity and reinitiation of growth associated with MMP9 activity. These findings underscore the importance of the dynamic reciprocity between the extracellular matrix integrity, tissue polarity, and Raf/MEK/ERK and MMP9 activities, providing an axis for either tissue homeostasis or malignant progression.


Asunto(s)
Neoplasias de la Mama/patología , Mama/citología , Metaloproteinasa 9 de la Matriz/metabolismo , Morfogénesis , Quinasas raf/fisiología , Animales , Técnicas de Cultivo de Célula , Polaridad Celular , Proliferación Celular , Humanos , Laminina/metabolismo , Ratones , Neoplasias Experimentales , Transducción de Señal , Trasplante Heterólogo
2.
Aging Cell ; 9(5): 736-46, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20569236

RESUMEN

The cyclin-dependent kinase inhibitor p16(INK4a) (CDKN2A) is an important tumor suppressor gene frequently inactivated in human tumors. p16 suppresses the development of cancer by triggering an irreversible arrest of cell proliferation termed cellular senescence. Here, we describe another anti-oncogenic function of p16 in addition to its ability to halt cell cycle progression. We show that transient expression of p16 stably represses the hTERT gene, encoding the catalytic subunit of telomerase, in both normal and malignant breast epithelial cells. Short-term p16 expression increases the amount of histone H3 trimethylated on lysine 27 (H3K27) bound to the hTERT promoter, resulting in transcriptional silencing, likely mediated by polycomb complexes. Our results indicate that transient p16 exposure may prevent malignant progression in dividing cells by irreversible repression of genes, such as hTERT, whose activity is necessary for extensive self-renewal.


Asunto(s)
Neoplasias de la Mama/enzimología , Mama/enzimología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Telomerasa/antagonistas & inhibidores , Mama/citología , Mama/metabolismo , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Proliferación Celular , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Células Epiteliales/citología , Células Epiteliales/enzimología , Silenciador del Gen , Histonas/metabolismo , Humanos , Metilación , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Telomerasa/genética , Telomerasa/metabolismo
3.
Cell Cycle ; 8(20): 3373-8, 2009 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-19806010

RESUMEN

A central question in breast cancer biology is how cancer cells acquire telomerase activity required for unlimited proliferation. According to one model, proliferation of telomerase(-) pre-malignant cells leads to telomere dysfunction and increased genomic instability. Such instability leads in rare cases to reactivation of telomerase and immortalization. The mechanism of telomerase reactivation remains unknown. We have studied immortalization of cultured human mammary epithelial cells by c-Myc, a positive transcriptional regulator of the hTERT gene encoding the catalytic subunit of telomerase. Retrovirally introduced c-Myc cDNA resulted in immortalization of human mammary epithelial cells in which the cyclin dependent kinase inhibitor, p16(INK4A), was inactivated by an shRNA-encoding retrovirus. However, while c-Myc introduction immediately resulted in increased activity of transiently transfected hTERT promoter reporter constructs, endogenous hTERT mRNA levels did not change until about 60 population doublings after c-Myc introduction. Increased endogenous hTERT transcripts and stabilization of telomeric DNA in cells expressing exogenous c-Myc coincided with telomere dysfunction-associated senescence in control cultures. Genome copy number analyses of immortalized cells indicated amplifications of some or all of chromosome 5, where hTERT genes are located. hTERT gene copy number, however, was not increased in one case. The results are consistent with the hypothesis that changes in chromosome 5, while not necessarily increasing hTERT gene copy number, resulted in removal of repressive chromatin structures around hTERT loci, allowing induction of hTERT transcription. These in vitro results model one possible sequence of events leading to immortalization of breast epithelial cells during cancer progression.


Asunto(s)
Células Epiteliales/metabolismo , Glándulas Mamarias Humanas/citología , Proteínas Proto-Oncogénicas c-myc/metabolismo , Telomerasa/metabolismo , Línea Celular Transformada , Cromosomas Humanos Par 5 , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Células Epiteliales/enzimología , Inestabilidad Genómica , Humanos , Proteínas Proto-Oncogénicas c-myc/genética , Interferencia de ARN , Telomerasa/genética
4.
Cell Cycle ; 6(11): 1284-7, 2007 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-17534147

RESUMEN

In addition to conferring an indefinite replicative life span, telomerase renders p16(-) human mammary epithelial cells (HMEC) resistant to growth arrest by TGFbeta or by loss of EGF or insulin signaling. In contrast to earlier reports, we recently found that growth factor signaling was not directly affected by telomerase expression. Rather, short dysfunctional or near-dysfunctional telomeres in proliferating telomerase(-) HMEC sensitized the cells to p53-dependent signals for growth arrest. We showed that during serial passage and before any signs of replicative senescence, HMEC lacking telomerase experience enhanced p53 stability and DNA damage signaling, as determined by increased phosphorylation on p53-Ser15 and Chk2-Thr68, and formation of 53BP1/phosphorylated histone H2AX foci at chromosome ends. This heightened activity of the p53 pathway enhanced the efficiency with which cells arrested growth in response to TGFbeta or to EGF or insulin withdrawal, and was abolished by ectopic expression of hTERT, the catalytic subunit of telomerase. Telomerase elongated short telomeres, thereby reducing the basal level of activated p53 and raising cellular tolerance for other p53-dependent signals, including those emanating from non-genotoxic sources. These findings explain a number of observed effects of telomerase expression on cell growth and survival without postulating additional functions for telomerase.


Asunto(s)
Estrés Fisiológico/genética , Telomerasa/fisiología , Proteína p53 Supresora de Tumor/fisiología , Animales , Mama/citología , División Celular , Línea Celular/citología , Línea Celular/efectos de los fármacos , Línea Celular/metabolismo , Senescencia Celular , Quinasa de Punto de Control 2 , Daño del ADN , Factor de Crecimiento Epidérmico/farmacología , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Humanos , Insulina/farmacología , Ratones , Fosforilación , Procesamiento Proteico-Postraduccional , Proteínas Serina-Treonina Quinasas/fisiología , Proteínas Recombinantes de Fusión/fisiología , Telomerasa/deficiencia , Telomerasa/genética , Telómero/metabolismo , Telómero/ultraestructura , Factor de Crecimiento Transformador beta/farmacología
5.
Proc Natl Acad Sci U S A ; 104(11): 4431-6, 2007 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-17360541

RESUMEN

Ectopically expressed hTERT enables p16(INK4A)(-) human mammary epithelial cells to proliferate in the absence of growth factors, a finding that has led to the hypothesis that hTERT has growth regulatory properties independent of its role in telomere maintenance. We now show that telomerase can alter the growth properties of cells indirectly through its role in telomere maintenance, without altering growth stimulatory pathways. We find that telomere dysfunction, indicated by 53BP1/phosphorylated histone H2AX foci at chromosome ends, is present in robustly proliferating human mammary epithelial cells long before senescence. These foci correlate with increased levels of active p53. Ectopic expression of hTERT reduces the number of foci and the level of active p53, thereby decreasing sensitivity to growth factor depletion, which independently activates p53. The continuous presence of hTERT is not necessary for this effect, indicating that telomere maintenance, rather than the presence of the enzyme itself, is responsible for the increased ability to proliferate in the absence of growth factors. Our findings provide a previously unrecognized mechanistic explanation for the observation that ectopically expressed hTERT conveys growth advantages to cells, without having to postulate nontelomeric functions for the enzyme.


Asunto(s)
Histonas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Transducción de Señal , Telómero/ultraestructura , Proteína p53 Supresora de Tumor/metabolismo , Proliferación Celular , Senescencia Celular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Daño del ADN , Factor de Crecimiento Epidérmico/metabolismo , Histonas/química , Humanos , Insulina/metabolismo , Glándulas Mamarias Humanas/citología , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA