Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Genes Dev ; 36(9-10): 511-513, 2022 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-35680423

RESUMEN

Senescence is a specialized form of cell cycle arrest induced in response to damage and stress. In certain settings, senescent cells can promote their own removal by recruitment of the immune system, a process that is thought to decline in efficiency with age. In this issue of Genes & Development, Yin et al. (pp. 533-549) discover a surprising cross-talk where senescent cells instruct endothelial cells to help organize the clearance of the senescent population. This uncovers yet another layer of complexity in senescent cell biology, with implications for cancer treatment and aging.


Asunto(s)
Senescencia Celular , Células Endoteliales , Puntos de Control del Ciclo Celular , Senescencia Celular/genética
2.
Cell ; 155(5): 1119-30, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24238961

RESUMEN

Senescence is a form of cell-cycle arrest linked to tumor suppression and aging. However, it remains controversial and has not been documented in nonpathologic states. Here we describe senescence as a normal developmental mechanism found throughout the embryo, including the apical ectodermal ridge (AER) and the neural roof plate, two signaling centers in embryonic patterning. Embryonic senescent cells are nonproliferative and share features with oncogene-induced senescence (OIS), including expression of p21, p15, and mediators of the senescence-associated secretory phenotype (SASP). Interestingly, mice deficient in p21 have defects in embryonic senescence, AER maintenance, and patterning. Surprisingly, the underlying mesenchyme was identified as a source for senescence instruction in the AER, whereas the ultimate fate of these senescent cells is apoptosis and macrophage-mediated clearance. We propose that senescence is a normal programmed mechanism that plays instructive roles in development, and that OIS is an evolutionarily adapted reactivation of a developmental process.


Asunto(s)
Senescencia Celular , Desarrollo Embrionario , Animales , Apoptosis , Embrión de Pollo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/inmunología , Embrión de Mamíferos/metabolismo , Extremidades/embriología , Fibroblastos/citología , Humanos , Ratones , Comunicación Paracrina
3.
Genes Dev ; 34(7-8): 489-494, 2020 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-32139422

RESUMEN

Young mammals possess a limited regenerative capacity in some tissues, which is lost upon maturation. We investigated whether cellular senescence might play a role in such loss during liver regeneration. We found that following partial hepatectomy, the senescence-associated genes p21, p16Ink4a, and p19Arf become dynamically expressed in different cell types when regenerative capacity decreases, but without a full senescent response. However, we show that treatment with a senescence-inhibiting drug improves regeneration, by disrupting aberrantly prolonged p21 expression. This work suggests that senescence may initially develop from heterogeneous cellular responses, and that senotherapeutic drugs might be useful in promoting organ regeneration.


Asunto(s)
Compuestos de Bifenilo/farmacología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Regulación de la Expresión Génica/efectos de los fármacos , Hígado/fisiología , Nitrofenoles/farmacología , Regeneración/efectos de los fármacos , Sulfonamidas/farmacología , Animales , Células Cultivadas , Senescencia Celular/efectos de los fármacos , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Piperazinas/farmacología
4.
PLoS Biol ; 20(6): e3001664, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35700169

RESUMEN

Valproic acid (VPA) is a widely prescribed drug to treat epilepsy, bipolar disorder, and migraine. If taken during pregnancy, however, exposure to the developing embryo can cause birth defects, cognitive impairment, and autism spectrum disorder. How VPA causes these developmental defects remains unknown. We used embryonic mice and human organoids to model key features of VPA drug exposure, including exencephaly, microcephaly, and spinal defects. In the malformed tissues, in which neurogenesis is defective, we find pronounced induction of cellular senescence in the neuroepithelial (NE) cells. Critically, through genetic and functional studies, we identified p19Arf as the instrumental mediator of senescence and microcephaly, but, surprisingly, not exencephaly and spinal defects. Together, these findings demonstrate that misregulated senescence in NE cells can contribute to developmental defects.


Asunto(s)
Trastorno del Espectro Autista , Microcefalia , Defectos del Tubo Neural , Animales , Senescencia Celular , Femenino , Ratones , Embarazo , Ácido Valproico/farmacología
5.
Genes Dev ; 31(2): 172-183, 2017 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-28143833

RESUMEN

Senescence is a form of cell cycle arrest induced by stress such as DNA damage and oncogenes. However, while arrested, senescent cells secrete a variety of proteins collectively known as the senescence-associated secretory phenotype (SASP), which can reinforce the arrest and induce senescence in a paracrine manner. However, the SASP has also been shown to favor embryonic development, wound healing, and even tumor growth, suggesting more complex physiological roles than currently understood. Here we uncover timely new functions of the SASP in promoting a proregenerative response through the induction of cell plasticity and stemness. We show that primary mouse keratinocytes transiently exposed to the SASP exhibit increased expression of stem cell markers and regenerative capacity in vivo. However, prolonged exposure to the SASP causes a subsequent cell-intrinsic senescence arrest to counter the continued regenerative stimuli. Finally, by inducing senescence in single cells in vivo in the liver, we demonstrate that this activates tissue-specific expression of stem cell markers. Together, this work uncovers a primary and beneficial role for the SASP in promoting cell plasticity and tissue regeneration and introduces the concept that transient therapeutic delivery of senescent cells could be harnessed to drive tissue regeneration.


Asunto(s)
Plasticidad de la Célula/fisiología , Senescencia Celular/fisiología , Regeneración/fisiología , Vías Secretoras/fisiología , Animales , Biomarcadores/metabolismo , Plasticidad de la Célula/genética , Células Cultivadas , Senescencia Celular/genética , Células Epiteliales/citología , Células Epiteliales/fisiología , Femenino , Eliminación de Gen , Regulación del Desarrollo de la Expresión Génica/genética , Queratinocitos/citología , Queratinocitos/fisiología , Hígado/citología , Hígado/fisiología , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , Fenotipo , Regeneración/genética , Vías Secretoras/genética , Células Madre/metabolismo
6.
Development ; 146(20)2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31575608

RESUMEN

Cellular senescence is a state comprising an essentially irreversible proliferative arrest combined with phenotypic changes and pronounced secretory activity. Although senescence has long been linked with aging, recent studies have uncovered functional roles for senescence in embryonic development, regeneration and reprogramming, and have helped to advance our understanding of this process as a highly coordinated and programmed cellular state. In this Primer article, we summarize some of the key findings in the field and attempt to explain them in a simple model that reconciles the normal and pathological roles for senescence. We discuss how a primary role of cellular senescence is to contribute to normal development, cell plasticity and tissue repair, as a dynamic and tightly regulated cellular program. However, when this process is perturbed, the beneficial effects turn detrimental and can contribute to disease and aging.


Asunto(s)
Senescencia Celular/fisiología , Regeneración/fisiología , Envejecimiento/genética , Envejecimiento/fisiología , Animales , Plasticidad de la Célula/genética , Plasticidad de la Célula/fisiología , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Senescencia Celular/genética , Humanos , Regeneración/genética
7.
Genes Dev ; 26(19): 2144-53, 2012 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-22972935

RESUMEN

Altered stem cell homeostasis is linked to organismal aging. However, the mechanisms involved remain poorly understood. Here we report novel alterations in hair follicle stem cells during skin aging, including increased numbers, decreased function, and an inability to tolerate stress. Performing high-throughput RNA sequencing on aging stem cells, cytokine arrays, and functional assays, we identify an age-associated imbalance in epidermal Jak-Stat signaling that inhibits stem cell function. Collectively, this study reveals a role for the aging epidermis in the disruption of cytokine and stem cell homeostasis, suggesting that stem cell decline during aging may be part of broader tumor-suppressive mechanisms.


Asunto(s)
Envejecimiento , Células Epidérmicas , Inflamación , Células Madre/citología , Animales , Recuento de Células , Células Cultivadas , Citocinas/metabolismo , Epidermis/enzimología , Folículo Piloso/citología , Folículo Piloso/enzimología , Homeostasis/fisiología , Quinasas Janus/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Células Madre/enzimología
8.
EMBO J ; 32(16): 2217-30, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23872946

RESUMEN

Cellular senescence is an intrinsic defense mechanism to various cellular stresses: while still metabolically active, senescent cells stop dividing and enter a proliferation arrest. Here, we identify DPY30, a member of all mammalian histone H3K4 histone methyltransferases (HMTases), as a key regulator of the proliferation potential of human primary cells. Following depletion of DPY30, cells show a severe proliferation defect and display a senescent phenotype, including a flattened and enlarged morphology, elevated level of reactive oxygen species (ROS), increased SA-ß-galactosidase activity, and formation of senescence-associated heterochromatin foci (SAHFs). While DPY30 depletion leads to a reduced level of H3K4me3-marked active chromatin, we observed a concomitant activation of CDK inhibitors, including p16INK4a, independent of H3K4me3. ChIP experiments show that key regulators of cell-cycle progression, including ID proteins, are under direct control of DPY30. Because ID proteins are negative regulators of the transcription factors ETS1/2, depletion of DPY30 leads to the transcriptional activation of p16INK4a by ETS1/2 and thus to a senescent-like phenotype. Ectoptic re-introduction of ID protein expression can partially rescue the senescence-like phenotype induced by DPY30 depletion. Thus, our data indicate that DPY30 controls proliferation by regulating ID proteins expression, which in turn lead to senescence bypass.


Asunto(s)
Senescencia Celular/fisiología , Regulación de la Expresión Génica/fisiología , Proteína 1 Inhibidora de la Diferenciación/metabolismo , Proteínas Nucleares/metabolismo , Transducción de Señal/fisiología , Western Blotting , Inmunoprecipitación de Cromatina , Ensayo de Unidades Formadoras de Colonias , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Análisis por Micromatrices , Proteínas Nucleares/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción , beta-Galactosidasa
9.
Biochem Soc Trans ; 42(3): 663-9, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24849235

RESUMEN

Adult tissue homoeostasis requires continual replacement of cells that are lost due to normal turnover, injury and disease. However, aging is associated with an overall decline in tissue function and homoeostasis, suggesting that the normal regulatory processes that govern self-renewal and regeneration may become impaired with age. Tissue-specific SCs (stem cells) lie at the apex of organismal conservation and regeneration, ultimately being responsible for continued tissue maintenance. In many tissues, there are changes in SC numbers, or alteration of their growth properties during aging, often involving imbalances in tumour-suppressor- and oncogene-mediated pathways. Uncovering the molecular mechanisms leading to changes in SC function during aging will provide an essential tool to address tissue-specific age-related pathologies. In the present review, we summarize the age-related alterations found in different tissue SC populations, highlighting recently identified changes in aged HFSCs (hair-follicle SCs) in the skin.


Asunto(s)
Senescencia Celular , Neoplasias/patología , Piel/citología , Células Madre/citología , Compartimento Celular , Humanos
10.
FEBS J ; 290(5): 1303-1313, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36856681

RESUMEN

Cellular senescence is a distinct state that is frequently induced in response to ageing and stress. Yet studies have also uncovered beneficial functions in development, repair and regeneration. Current opinion therefore suggests that timely and controlled induction of senescence can be beneficial, while misregulation of the senescence program, either through mis-timed activation, or chronic accumulation of senescent cells, contributes to many disease states and the ageing process. Whether atypical activation of senescence plays a role in the pathogenesis of developmental defects has been relatively underexplored. Here, we discuss three recent studies that implicate ectopic senescence in neurodevelopmental defects, with possible causative roles for senescence in these birth defects. In addition, we highlight how the examination of senescence in other birth defects is warranted, and speculate that aberrantly activated senescence may play a much broader role in developmental defects than currently appreciated.


Asunto(s)
Senescencia Celular , Anomalías Congénitas
11.
Nat Commun ; 13(1): 2865, 2022 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-35606383

RESUMEN

Despite the fact that the cell cycle is a fundamental process of life, a detailed quantitative understanding of gene regulation dynamics throughout the cell cycle is far from complete. Single-cell RNA-sequencing (scRNA-seq) technology gives access to these dynamics without externally perturbing the cell. Here, by generating scRNA-seq libraries in different cell systems, we observe cycling patterns in the unspliced-spliced RNA space of cell cycle-related genes. Since existing methods to analyze scRNA-seq are not efficient to measure cycling gene dynamics, we propose a deep learning approach (DeepCycle) to fit these patterns and build a high-resolution map of the entire cell cycle transcriptome. Characterizing the cell cycle in embryonic and somatic cells, we identify major waves of transcription during the G1 phase and systematically study the stages of the cell cycle. Our work will facilitate the study of the cell cycle in multiple cellular models and different biological contexts.


Asunto(s)
Aprendizaje Profundo , Análisis de la Célula Individual , Perfilación de la Expresión Génica/métodos , Genes cdc , ARN/genética , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Transcriptoma
13.
Neuron ; 48(5): 743-56, 2005 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-16337913

RESUMEN

The p53 family member p63 is required for nonneural development, but has no known role in the nervous system. Here, we define an essential proapoptotic role for p63 during naturally occurring neuronal death. Sympathetic neurons express full-length TAp63 during the developmental death period, and TAp63 levels increase following NGF withdrawal. Overexpression of TAp63 causes neuronal apoptosis in the presence of NGF, while cultured p63-/- neurons are resistant to apoptosis following NGF withdrawal. TAp63 is also essential in vivo, since embryonic p63-/- mice display a deficit in naturally occurring sympathetic neuron death. While both TAp63 and p53 induce similar apoptotic signaling proteins and require BAX expression and function for their effects, TAp63 induces neuronal death in the absence of p53, but p53 requires coincident p63 expression for its proapoptotic actions. Thus, p63 is essential for developmental neuronal death, likely functioning both on its own, and as an obligate proapoptotic partner for p53.


Asunto(s)
Apoptosis/fisiología , Fosfoproteínas/fisiología , Sistema Nervioso Simpático/embriología , Sistema Nervioso Simpático/crecimiento & desarrollo , Transactivadores/fisiología , Envejecimiento/fisiología , Animales , Animales Recién Nacidos , Muerte Celular/fisiología , Células Cultivadas , Proteínas Quinasas JNK Activadas por Mitógenos/fisiología , Ratones , Ratones Endogámicos , Ratones Noqueados , Ratones Mutantes , Mitocondrias/fisiología , Factor de Crecimiento Nervioso/administración & dosificación , Factor de Crecimiento Nervioso/farmacología , Neuronas/metabolismo , Neuronas/fisiología , Fosfoproteínas/deficiencia , Fosfoproteínas/metabolismo , Isoformas de Proteínas/metabolismo , Ratas , Ratas Sprague-Dawley , Sistema Nervioso Simpático/citología , Transactivadores/deficiencia , Transactivadores/metabolismo , Proteína p53 Supresora de Tumor/fisiología , Proteína X Asociada a bcl-2/fisiología
14.
Methods Mol Biol ; 1534: 199-210, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-27812881

RESUMEN

Senescence-associated ß-galactosidase (SAß-gal) is a convenient histological technique used to identify senescent cells. Its ease of use is helpful to initially screen and detect senescent cells in heterogeneous cell populations both in vitro and in vivo. However, SAß-gal staining is not an unequivocal marker of the senescent state, and diagnosis of such usually requires additional markers demonstrating an absence of proliferation and expression of cell-cycle inhibitors. Nonetheless, SAß-gal remains one of the most widely used biomarkers of senescent cells. Recently, by measuring SAß-gal activity, the expression of the cyclin-dependent kinase inhibitor p21 (waf1/cip1) and demonstrating a lack of proliferation, we identified senescent cells in the developing embryo. This chapter describes the methods for identifying cellular senescence in the embryo, detailing protocols for the detection of SAß-gal activity in both sections and at the whole mount level, and immunohistochemistry protocols for the detection of additional biomarkers of senescence.


Asunto(s)
Biomarcadores , Senescencia Celular , Desarrollo Embrionario , Animales , Senescencia Celular/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/metabolismo , Desarrollo Embrionario/genética , Humanos , Inmunohistoquímica/métodos , Mamíferos , Ratones , beta-Galactosidasa/metabolismo
15.
Nat Aging ; 2(7): 570-572, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-37117776
16.
Trends Biotechnol ; 21(2): 53-5, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12573850
17.
Anat Embryol (Berl) ; 207(2): 135-47, 2003 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12905017

RESUMEN

Bone morphogenetic proteins (BMPs) are considered to be significant factors in the morphogenesis of the endocardial cushions of the developing embryonic heart. Previous studies have suggested that they are involved in the epithelial-mesenchymal transformation and migration of the cells forming the cushions, or in triggering an apoptotic cascade in a sub-population of cushion cells. We investigated the expression and function of BMP2 and BMP4 proteins in the developing heart of the chick and mouse embryos. In the chick, by immunocytochemistry, we find expression of BMP2 protein in the endocardial cushions of the outflow tract (OT) and atrio-ventricular (AV) regions at embryonic days (ED) 5-6, as well as in adjacent myocardial layers. Immunoblotting indicated that such expression persisted through ED 4-7, but peaked at ED4-5 in the OT and 5-6 in the AV cushions. This temporal sequence correlated with the peaks of apoptotic cell death found previously in the OT and AV cushions of the chick embryo. At equivalent stages in mouse, no such expression of BMP2 was found in the cushions, although expression was detected in adjacent myocardial layers. In the case of BMP4, in both chick and mouse, expression was found only in the myocardia and not in the cushions. Furthermore, BMP-specific receptors were found in the cushions, but not the myocardia, in both the AV and OT regions of the chick embryo. These results provide circumstantial evidence to support the contention that BMPs, originating from the myocardium, could be significant in the induction of apoptosis in chick embryo cushion cells, and confirms that there is species-specific variation in the expression pattern of BMP proteins, as had been predicted from previous studies of mRNA expression. Culture media conditioned by the growth of tissues from various regions of the developing heart were tested for their ability to induce apoptosis in cushion cells in culture. It was found that medium derived from the myocardia induced significant levels of cell death in the cushion cells, and that BMP4 could be detected in such media; however, retroviral over-expression of constitutively active (CA) and dominant-negative (DN) isoforms of BMP-specific receptors 1A and 1B (BMPR-1A and BMPR-1B) in cultured cells of the AV cushions did not alter levels of apoptosis or cell proliferation. Similar over-expression in cultured endocardial cells resulted in a significant change in cell shape, from endothelial to fibroblastic, with BMPR-1A CA and BMPR-1B DN, suggesting an influence of these receptors on cell transformation and/or cell migration. Taken together, these results provide support for the contention that BMP2 and BMP4 are important factors in the phenotypic transformational events involved in the morphogenesis of the chick embryo endocardial cushions, and could be involved in the induction of apoptosis in the cushion cells.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Endocardio/metabolismo , Factor de Crecimiento Transformador beta , Animales , Apoptosis/efectos de los fármacos , Proteína Morfogenética Ósea 2 , Proteína Morfogenética Ósea 4 , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1 , Tamaño de la Célula , Células Cultivadas , Embrión de Pollo , Medios de Cultivo Condicionados/farmacología , Desarrollo Embrionario y Fetal , Endocardio/efectos de los fármacos , Endocardio/embriología , Regulación Viral de la Expresión Génica , Edad Gestacional , Procesamiento de Imagen Asistido por Computador , Immunoblotting , Técnicas para Inmunoenzimas , Etiquetado Corte-Fin in Situ , Ratones , Proteínas Serina-Treonina Quinasas/metabolismo , Receptores de Factores de Crecimiento/metabolismo , Retroviridae/genética
18.
Commun Integr Biol ; 7(5)2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26842300

RESUMEN

Cellular senescence is an irreversible form of cell cycle arrest that has been linked to several pathological conditions. In particular, senescence can function as a tumor suppressor mechanism, but is also thought to contribute to organismal aging. Paradoxically however, through the secretion of various factors, collectively termed the senescence-associated secretory phenotype (SASP), senescent cells can also have tumor-promoting and tissue-remodeling functions. In addition, senescent cells can play beneficial roles in tissue repair and wound healing, and reconciling these contradictory features from an evolutionary standpoint has been challenging. Moreover, senescent cells had not previously been documented in non-pathological conditions. Recently however, 2 studies have identified cellular senescence as a programmed mechanism that contributes to tissue patterning and remodeling during normal embryonic development. These findings have significant implications for our understanding of cellular senescence and help to clarify the paradoxes and the evolutionary origin of this process.

19.
Cancer Cell ; 24(2): 151-66, 2013 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-23850221

RESUMEN

IκB proteins are the primary inhibitors of NF-κB. Here, we demonstrate that sumoylated and phosphorylated IκBα accumulates in the nucleus of keratinocytes and interacts with histones H2A and H4 at the regulatory region of HOX and IRX genes. Chromatin-bound IκBα modulates Polycomb recruitment and imparts their competence to be activated by TNFα. Mutations in the Drosophila IκBα gene cactus enhance the homeotic phenotype of Polycomb mutants, which is not counteracted by mutations in dorsal/NF-κB. Oncogenic transformation of keratinocytes results in cytoplasmic IκBα translocation associated with a massive activation of Hox. Accumulation of cytoplasmic IκBα was found in squamous cell carcinoma (SCC) associated with IKK activation and HOX upregulation.


Asunto(s)
Cromatina/metabolismo , Proteínas I-kappa B/metabolismo , Neoplasias Cutáneas/metabolismo , Animales , Diferenciación Celular/genética , Núcleo Celular/genética , Núcleo Celular/metabolismo , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Cromatina/genética , Células HEK293 , Histonas/genética , Histonas/metabolismo , Humanos , Proteínas I-kappa B/genética , Queratinocitos/citología , Queratinocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Inhibidor NF-kappaB alfa , Transducción de Señal , Neoplasias Cutáneas/genética , Neoplasias Cutáneas/patología
20.
Cell Stem Cell ; 8(2): 164-76, 2011 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-21295273

RESUMEN

The p53 homolog p63 is essential for development, yet its role in cancer is not clear. We discovered that p63 deficiency evokes the tumor-suppressive mechanism of cellular senescence, causing a striking absence of stratified epithelia such as the skin. Here we identify the predominant p63 isoform, ΔNp63α, as a protein that bypasses oncogene-induced senescence to drive tumorigenesis in vivo. Interestingly, bypass of senescence promotes stem-like proliferation and maintains survival of the keratin 15-positive stem cell population. Furthermore, we identify the chromatin-remodeling protein Lsh as a new target of ΔNp63α that is an essential mediator of senescence bypass. These findings indicate that ΔNp63α is an oncogene that cooperates with Ras to promote tumor-initiating stem-like proliferation and suggest that Lsh-mediated chromatin-remodeling events are critical to this process.


Asunto(s)
ADN Helicasas/metabolismo , Fosfoproteínas/metabolismo , Piel/citología , Células Madre/citología , Células Madre/metabolismo , Transactivadores/metabolismo , Animales , Proliferación Celular , Células Cultivadas , Inmunoprecipitación de Cromatina , Citometría de Flujo , Humanos , Queratinocitos/metabolismo , Ratones , Ratones Desnudos , Fosfoproteínas/genética , Reacción en Cadena de la Polimerasa , Unión Proteica , Transactivadores/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA