Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
EMBO J ; 38(11)2019 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-31000523

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are immature myeloid cells with strong immunosuppressive activity that promote tumor growth. In this study, we describe a mechanism by which cancer cells control MDSCs in human cancers by upregulating TRF2, a protein required for telomere stability. Specifically, we showed that the TRF2 upregulation in cancer cells has extratelomeric roles in activating the expression of a network of genes involved in the biosynthesis of heparan sulfate proteoglycan, leading to profound changes in glycocalyx length and stiffness, as revealed by atomic force microscopy. This TRF2-dependent regulation facilitated the recruitment of MDSCs, their activation via the TLR2/MyD88/IL-6/STAT3 pathway leading to the inhibition of natural killer recruitment and cytotoxicity, and ultimately tumor progression and metastasis. The clinical relevance of these findings is supported by our analysis of cancer cohorts, which showed a correlation between high TRF2 expression and MDSC infiltration, which was inversely correlated with overall patient survival.


Asunto(s)
Glicocálix/metabolismo , Neoplasias/inmunología , Neoplasias/patología , Proteína 2 de Unión a Repeticiones Teloméricas/fisiología , Escape del Tumor/fisiología , Animales , Células Cultivadas , Femenino , Regulación Neoplásica de la Expresión Génica , Glicocálix/genética , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Células Supresoras de Origen Mieloide/metabolismo , Células Supresoras de Origen Mieloide/fisiología , Células 3T3 NIH , Neoplasias/genética , Neoplasias/mortalidad , Telómero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Escape del Tumor/genética
2.
Nat Rev Genet ; 15(7): 491-503, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24913665

RESUMEN

Telomeres protect chromosome ends from degradation and inappropriate DNA damage response activation through their association with specific factors. Interestingly, these telomeric factors are able to localize outside telomeric regions, where they can regulate the transcription of genes involved in metabolism, immunity and differentiation. These findings delineate a signalling pathway by which telomeric changes control the ability of their associated factors to regulate transcription. This mechanism is expected to enable a greater diversity of cellular responses that are adapted to specific cell types and telomeric changes, and may therefore represent a pivotal aspect of development, ageing and telomere-mediated diseases.


Asunto(s)
Reparación del ADN , Transducción de Señal/genética , Telómero/química , Transcripción Genética , Apoptosis , Senescencia Celular , Daño del ADN , Regulación de la Expresión Génica , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Complejo Shelterina , Telomerasa/genética , Telomerasa/metabolismo , Telómero/metabolismo , Proteínas de Unión a Telómeros/genética , Proteínas de Unión a Telómeros/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
3.
EMBO Rep ; 14(4): 356-63, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23429341

RESUMEN

The DNA-binding protein TRF2 is essential for telomere protection and chromosome stability in mammals. We show here that TRF2 expression is activated by the Wnt/ß-catenin signalling pathway in human cancer and normal cells as well as in mouse intestinal tissues. Furthermore, ß-catenin binds to TRF2 gene regulatory regions that are functional in a luciferase transactivating assay. Reduced ß-catenin expression in cancer cells triggers a marked increase in telomere dysfunction, which can be reversed by TRF2 overexpression. We conclude that the Wnt/ß-catenin signalling pathway maintains a level of TRF2 critical for telomere protection. This is expected to have an important role during development, adult stem cell function and oncogenesis.


Asunto(s)
Regulación de la Expresión Génica , Homeostasis del Telómero , Proteína 2 de Unión a Repeticiones Teloméricas/metabolismo , Vía de Señalización Wnt , Animales , Sitios de Unión , Femenino , Expresión Génica , Células HCT116 , Humanos , Masculino , Ratones , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Unión Proteica , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteína 2 de Unión a Repeticiones Teloméricas/genética , Transcriptoma , beta Catenina/metabolismo
4.
Genome Res ; 21(5): 798-810, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21451113

RESUMEN

Emerging evidence suggests that microRNAs (miRNAs), an abundant class of ∼22-nucleotide small regulatory RNAs, play key roles in controlling the post-transcriptional genetic programs in stem and progenitor cells. Here we systematically examined miRNA expression profiles in various adult tissue-specific stem cells and their differentiated counterparts. These analyses revealed miRNA programs that are common or unique to blood, muscle, and neural stem cell populations and miRNA signatures that mark the transitions from self-renewing and quiescent stem cells to proliferative and differentiating progenitor cells. Moreover, we identified a stem/progenitor transition miRNA (SPT-miRNA) signature that predicts the effects of genetic perturbations, such as loss of PTEN and the Rb family, AML1-ETO9a expression, and MLL-AF10 transformation, on self-renewal and proliferation potentials of mutant stem/progenitor cells. We showed that some of the SPT-miRNAs control the self-renewal of embryonic stem cells and the reconstitution potential of hematopoietic stem cells (HSCs). Finally, we demonstrated that SPT-miRNAs coordinately regulate genes that are known to play roles in controlling HSC self-renewal, such as Hoxb6 and Hoxa4. Together, these analyses reveal the miRNA programs that may control key processes in normal and aberrant stem and progenitor cells, setting the foundations for dissecting post-transcriptional regulatory networks in stem cells.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/metabolismo , Regulación de la Expresión Génica/genética , MicroARNs/metabolismo , Células Madre/metabolismo , Animales , Diferenciación Celular/genética , Células Madre Embrionarias/citología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Humanos , Ratones , MicroARNs/genética , Mutación , Mioblastos/citología , Mioblastos/metabolismo , Células-Madre Neurales , Especificidad de Órganos , Células Madre/citología
5.
Aging (Albany NY) ; 3(2): 108-24, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21386132

RESUMEN

In adult mammals, neural stem cells (NSCs) generate new neurons that are important for specific types of learning and memory. Controlling adult NSC number and function is fundamental for preserving the stem cell pool and ensuring proper levels of neurogenesis throughout life. Here we study the importance of the microRNA gene cluster miR-106b~25 (miR-106b, miR-93, and miR-25) in primary cultures of neural stem/progenitor cells (NSPCs) isolated from adult mice. We find that knocking down miR-25 decreases NSPC proliferation, whereas ectopically expressing miR-25 promotes NSPC proliferation. Expressing the entire miR-106b~25 cluster in NSPCs also increases their ability to generate new neurons. Interestingly, miR-25 has a number of potential target mRNAs involved in insulin/insulin-like growth factor-1 (IGF) signaling, a pathway implicated in aging. Furthermore, the regulatory region of miR-106b~25 is bound by FoxO3, a member of the FoxO family of transcription factors that maintains adult stem cells and extends lifespan downstream of insulin/IGF signaling. These results suggest that miR-106b~25 regulates NSPC function and is part of a network involving the insulin/IGF-FoxO pathway, which may have important implications for the homeostasis of the NSC pool during aging.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular , MicroARNs/genética , Familia de Multigenes , Células-Madre Neurales/fisiología , Neurogénesis/fisiología , Animales , Células Cultivadas , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Insulina/metabolismo , Ratones , MicroARNs/metabolismo , Células-Madre Neurales/citología , Transducción de Señal/fisiología , Factor de Crecimiento Transformador beta/genética , Factor de Crecimiento Transformador beta/metabolismo
6.
Stem Cells Dev ; 20(7): 1233-46, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20954847

RESUMEN

Embryonic stem (ES) cells differentiate in vitro into all cell lineages. We previously found that the p38 mitogen activated kinase (p38MAPK) pathway controls the commitment of ES cells toward either cardiomyogenesis (p38 on) or neurogenesis (p38 off ). In this study, we show that p38α knock-out ES cells do not differentiate into cardiac, endothelial, smooth muscle, and skeletal muscle lineages. Reexpression of p38MAPK in these cells partially rescues their mesodermal differentiation defects and corrects the high level of spontaneous neurogenesis of knock-out cells. Wild-type ES cells were treated with a p38MAPK-specific inhibitor during the differentiation process. These experiments allowed us to identify 2 early independent successive p38MAPK functions in the formation of mesodermal lineages. Further, the first one correlates with the regulation of the expression of Brachyury, an essential mesodermal-specific transcription factor, by p38MAPK. In conclusion, by genetic and biochemical approaches, we demonstrate that p38MAPK activity is essential for the commitment of ES cell into cardiac, endothelial, smooth muscle, and skeletal muscle mesodermal lineages.


Asunto(s)
Diferenciación Celular , Células Madre Embrionarias/citología , Mesodermo/citología , Proteína Quinasa 14 Activada por Mitógenos/metabolismo , Animales , Western Blotting , Células Cultivadas , Células Madre Embrionarias/efectos de los fármacos , Células Madre Embrionarias/metabolismo , Proteínas Fetales/genética , Proteínas Fetales/metabolismo , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Imidazoles/farmacología , Mesodermo/metabolismo , Ratones , Proteína Quinasa 14 Activada por Mitógenos/antagonistas & inhibidores , Desarrollo de Músculos , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo
7.
Cell Stem Cell ; 5(5): 527-39, 2009 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-19896443

RESUMEN

In the nervous system, neural stem cells (NSCs) are necessary for the generation of new neurons and for cognitive function. Here we show that FoxO3, a member of a transcription factor family known to extend lifespan in invertebrates, regulates the NSC pool. We find that adult FoxO3(-/-) mice have fewer NSCs in vivo than wild-type counterparts. NSCs isolated from adult FoxO3(-/-) mice have decreased self-renewal and an impaired ability to generate different neural lineages. Identification of the FoxO3-dependent gene expression profile in NSCs suggests that FoxO3 regulates the NSC pool by inducing a program of genes that preserves quiescence, prevents premature differentiation, and controls oxygen metabolism. The ability of FoxO3 to prevent the premature depletion of NSCs might have important implications for counteracting brain aging in long-lived species.


Asunto(s)
Células Madre Adultas/metabolismo , Encéfalo/metabolismo , Factores de Transcripción Forkhead/metabolismo , Neuronas/metabolismo , Oxígeno/metabolismo , Células Madre Adultas/citología , Animales , Diferenciación Celular/genética , Linaje de la Célula/genética , Células Cultivadas , Proteína Forkhead Box O3 , Factores de Transcripción Forkhead/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Homeostasis , Ratones , Ratones Noqueados , Neurogénesis , Neuronas/citología
8.
Semin Cell Dev Biol ; 16(4-5): 612-22, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-16087370

RESUMEN

The Notch signaling pathway is an evolutionarily conserved pathway that is critical for tissue morphogenesis during development, but is also involved in tissue maintenance and repair in the adult. In skeletal muscle, regulation of Notch signaling is involved in somitogenesis, muscle development, and the proliferation and cell fate determination of muscle stems cells during regeneration. During each of these processes, the spatial and temporal control of Notch signaling is essential for proper tissue formation. That control is mediated by a series of regulatory proteins and protein complexes that enhance or inhibit Notch signaling by regulating protein processing, localization, activity, and stability. In this review, we focus on the regulation of Notch signaling during postnatal muscle regeneration when muscle stem cells ("satellite cells") must activate, proliferate, progress along a myogenic lineage pathway, and ultimately differentiate to form new muscle. We review the regulators of Notch signaling, such as Numb and Deltex, that have documented roles in myogenesis as well as other regulators that may play a role in modulating Notch signaling during satellite cell activation and postnatal myogenesis.


Asunto(s)
Animales Recién Nacidos/fisiología , Células Madre Mesenquimatosas/fisiología , Desarrollo de Músculos/fisiología , Receptores Notch/fisiología , Células Satélite del Músculo Esquelético/fisiología , Transducción de Señal/fisiología , Animales , Animales Recién Nacidos/metabolismo , Humanos , Receptores Notch/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA