Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Sci Rep ; 10(1): 7140, 2020 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-32346031

RESUMEN

The eukaryotic cell cycle is negatively regulated by cyclin-dependent kinase inhibitors (CKIs). p57Kip2 is a member of the Cip/Kip family of CKIs and frequently inactivated by genomic mutations associated with human overgrowth disorders. There is increasing evidence for p57 to control cellular processes in addition to cell cycle and CDK regulation including transcription, apoptosis, migration or development. In order to obtain molecular insights to unknown functions of p57, we performed a protein interaction screen. We identified the transcription regulator four-and-a-half LIM-only protein 2 (FHL2) as a novel p57-binding protein. Co-immunoprecipitation and reporter gene assays were used to elucidate the physiological and functional relevance of p57/FHL2 interaction. We found in cancer cells that endogenous p57 and FHL2 are in a complex. We observed a substantial induction of established FHL2-regulated gene promoters by p57 in reporter gene experiments and detected strong induction of the intrinsic transactivation activity of FHL2. Treatment of cells with histone deacetylase (HDAC) inhibitors and binding of exogenous FHL2 to HDACs indicated repression of FHL2 transcription activity by HDACs. In the presence of the HDAC inhibitor sodium butyrate activation of FHL2 by p57 is abrogated suggesting that p57 shares a common pathway with HDAC inhibitors. p57 competes with HDACs for FHL2 binding which might partly explain the mechanism of FHL2 activation by p57. These results suggest a novel function of p57 in transcription regulation.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Proteínas con Homeodominio LIM/fisiología , Proteínas Musculares/fisiología , Transactivadores/fisiología , Factores de Transcripción/fisiología , Ensamble y Desensamble de Cromatina , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Proteínas con Homeodominio LIM/metabolismo , Proteínas Musculares/metabolismo , Unión Proteica , Fracciones Subcelulares/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética
2.
Int J Mol Sci ; 19(9)2018 Sep 12.
Artículo en Inglés | MEDLINE | ID: mdl-30213134

RESUMEN

Cyclin dependent kinase inhibitor 1c (Cdkn1c) is a maternally expressed imprinted gene with roles in embryonic development, post-natal metabolism and behaviour. Using mouse models with altered dosages of Cdkn1c, we have previously identified a role for the gene in promoting brown adipose tissue formation. Here, we use these transgenic mouse lines to model the loss of imprinting of Cdkn1c in adulthood. We demonstrate that only a two-fold increase in the expression of Cdkn1c during development is sufficient to protect against age-related weight gain in addition to glucose and insulin intolerance. Further to this, we show that the loss of imprinting of Cdkn1c protects against diet-induced obesity. Bisulphite sequencing was performed to test the stability of the two differentially methylated regions that regulate Cdkn1c imprinting, and both were found to be unaltered in aged or diet-challenged adipose tissue, despite drastic reductions in Cdkn1c expression. These data demonstrate a critical role for Cdkn1c in regulating adult adipose tissue, with modest changes in expression capable of protecting against both age and diet-induced obesity and metabolic syndrome, with a natural decline in Cdkn1c expression observed that may contribute to less healthy metabolic aging. Finally, we have observed a post-natal insensitivity of the imprint to environmental factors, in contrast to recent observations of an in utero sensitivity.


Asunto(s)
Envejecimiento/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Impresión Genómica/genética , Obesidad/genética , Envejecimiento/fisiología , Animales , Cromosomas Artificiales Bacterianos , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Dieta Alta en Grasa/efectos adversos , Femenino , Masculino , Ratones , Obesidad/etiología , Obesidad/fisiopatología
3.
J Mol Histol ; 49(5): 459-469, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30014245

RESUMEN

Cdkn1c, a member of the Cip/Kip cyclin-dependent kinase inhibitor family, is critically involved in regulating cell cycle and cellular differentiation during development in mammals. However, the functional role of Cdkn1c and the underlying mechanisms by which Cdkn1c affects odontogenesis remain largely unknown. In our study, we found that Cdkn1c expression dynamically changes from embryonic day 11.5 (E11.5) to postnatal day 3 (P3), and exhibits tissue-specific expression profiles. Evaluation of CDKN1C protein by immunohistochemistry and western blot, revealed that CDKN1C protein expression peaks at P3 and then is reduced at P5 and P7. Interestingly, we observed that CDKN1C expression is higher in immature odontoblasts than preodontoblasts, is lower in mature odontoblasts, and is practically absent from ameloblasts. We evaluated cell cycle progression to further investigate the mechanisms underlying CDKN1C-mediated regulation of odontogenesis, and found that pRB, cyclin D1 and CDK2 expression decreased from P1 to P3, and reduced at P5 and P7. In addition, we observed increased methylation of KvDMR1 at P1 and P3, and reduced KvDMR1 methylation at P5 and P7. However, the methylation levels of Cdkn1c-sDMR were relatively low from P1 to P7. In summary, we demonstrated that Cdkn1c expression and methylation status may be involved in early postnatal tooth development through regulating the cell cycle inhibition activity of Cdkn1c. Notably, Cdkn1c expression and methylation may associate with cell cycle exit and differentiation of odontoblasts.


Asunto(s)
Ciclo Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Odontogénesis , Animales , Animales Recién Nacidos , Diferenciación Celular , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Metilación de ADN , Embrión de Mamíferos , Metilación , Ratones , Ratones Endogámicos BALB C , Odontoblastos/citología
4.
Front Immunol ; 9: 549, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29632530

RESUMEN

Transition from resting to cell cycle in response to antigenic stimulation is an essential step for naïve CD8+ T cells to differentiate to effector and memory cells. Leaving the resting state requires dramatic changes of chromatin status in the key cell cycle inhibitors but the details of these concerted events are not fully elucidated. Here, we showed that Ezh2, an enzymatic component of polycomb repressive complex 2 (PRC2) catalyzing the trimethylation of lysine 27 on histone 3 (H3K27me3), regulates activation induced naïve CD8+ T cells proliferation and apoptosis. Upon deletion of Ezh2 during thymocyte development (Ezh2fl/flCd4Cre+ mice), naive CD8+ T cells displayed impaired proliferation and increased apoptosis in response to antigen stimulation. However, naive CD8+ T cells only had impaired proliferation but no increase in apoptosis when Ezh2 was deleted after activation (Ezh2fl/flGzmBCre+ mice), suggesting cell cycle and apoptosis are temporally separable events controlled by Ezh2. We then showed that deletion of Ezh2 resulted in the increase in expression of cyclin-dependent kinase inhibitors Cdkn2a (p16 and Arf) and Cdkn1c (p57) in activated naïve CD8+ T cells as the consequence of reduced levels of H3K27me3 at these two gene loci. Finally, with real time imaging, we observed prolonged cell division times of naïve CD8+ T cells in the absence of Ezh2 post in vitro stimulation. Together, these findings reveal that repression of Cdkn1c and Cdkn2a by Ezh2 plays a critical role in execution of activation-induced CD8+ T cell proliferation.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Ciclo Celular/fisiología , Inhibidor p16 de la Quinasa Dependiente de Ciclina/fisiología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Proteína Potenciadora del Homólogo Zeste 2/fisiología , Animales , Antígenos/inmunología , Apoptosis , Proliferación Celular , Listeria monocytogenes , Listeriosis/inmunología , Ratones Noqueados , Ovalbúmina/inmunología
6.
Nat Neurosci ; 18(5): 657-65, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25821910

RESUMEN

The mechanism by which adult neural stem cells (NSCs) are established during development is unclear. In this study, analysis of cell cycle progression by examining retention of a histone 2B (H2B)-GFP fusion protein revealed that, in a subset of mouse embryonic neural progenitor cells (NPCs), the cell cycle slows between embryonic day (E) 13.5 and E15.5 while other embryonic NPCs continue to divide rapidly. By allowing H2B-GFP expressed at E9.5 to become diluted in dividing cells until the young adult stage, we determined that a majority of NSCs in the young adult subependymal zone (SEZ) originated from these slowly dividing embryonic NPCs. The cyclin-dependent kinase inhibitor p57 is highly expressed in this embryonic subpopulation, and the deletion of p57 impairs the emergence of adult NSCs. Our results suggest that a substantial fraction of adult SEZ NSCs is derived from a slowly dividing subpopulation of embryonic NPCs and identify p57 as a key factor in generating this embryonic origin of adult SEZ NSCs.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Proteínas del Tejido Nervioso/fisiología , Células-Madre Neurales/citología , Neurogénesis , Telencéfalo/citología , Animales , Ciclo Celular , División Celular , Linaje de la Célula , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Genes Reporteros , Histonas/biosíntesis , Histonas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos ICR , Proteínas del Tejido Nervioso/biosíntesis , Proteínas del Tejido Nervioso/genética , Proteínas Recombinantes de Fusión/metabolismo , Telencéfalo/embriología
7.
Oncogene ; 34(27): 3568-81, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25195859

RESUMEN

The DNA damage response (DDR) helps to maintain genome integrity, suppress tumorigenesis and mediate the radiotherapeutic and chemotherapeutic effects on cancer. Here we report that p57Kip2, a cyclin-dependent kinase (CDK) inhibitor implicated in the development of tumor-prone Beckwith-Wiedemann syndrome, is an effector molecule of the DNA-damage response. Genotoxic stress induces p57Kip2 expression via the bone morphogenetic protein-Smad1 and Atm-p38MAPK-Atf2 pathways in p53-proficient or -deficient cells and requires the Smad1-Atf2 complex that facilitates their recruitment to the p57Kip2 promoter. Elevated p57Kip2 induces G1/S phase cell cycle arrest but inhibits cell death in response to DNA damage and acts in parallel with p53 to suppress cell transformation and tumor formation. p57Kip2 is also upregulated in stage I and II clinical rectal tumor samples, likely due to genome instability of precancerous and/or early cancer cells. Targeting p57Kip2 in primary rectal cancer cells and tumor models resulted in increased sensitivity to doxorubicin, suggesting that p57Kip2 has a role in chemoresistance, which is consistent with its pro-survival function. These findings place p57Kip2 in DDR and uncover molecular mechanisms by which p57Kip2 suppresses tumorigenesis and causes chemoresistance.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Daño del ADN , Resistencia a Antineoplásicos/genética , Estrés Fisiológico/genética , Animales , Células Cultivadas , Daño del ADN/genética , Genes Supresores de Tumor , Células HCT116 , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Ratones Desnudos
8.
Cardiovasc Res ; 100(1): 95-104, 2013 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-23787002

RESUMEN

AIMS: Mammalian cardiomyogenesis occurs through a multistep process that requires a complex network of tightly regulated extracellular signals, which integrate with the genetic and epigenetic machinery to maintain, expand, and regulate the differentiation of cardiac progenitor cells. Pluripotent embryonic stem cells (ESCs) recapitulate many aspects of development, and have provided an excellent opportunity to dissect the molecular mechanisms underlying cardiomyogenesis, which is still incompletely defined. METHODS AND RESULTS: We provide new in vivo evidence that the G-protein-coupled receptor angiotensin receptor-like 1 (Apj) is expressed in the mesodermal cells of the second heart field, a population of cardiac progenitors that give rise to a major part of the definitive heart. By combining loss-and-gain of function studies in mouse ESCs, we show that Apj (i) controls the balance between proliferation and cardiovascular differentiation, (ii) regulates the Nodal/Bone Morphogenetic Protein antagonist Cerberus and the Baf60c/Smarcd3 subunit of the Brg1/Brm-associated factors (BAF) chromatin-remodelling complex. CONCLUSION: We propose a model in which Apj controls a regulatory Cerberus-Baf60c pathway in pluripotent stem cell cardiomyogenesis, and speculate that this regulatory circuit may regulate cardiac progenitor cell behaviour.


Asunto(s)
Proteínas Morfogenéticas Óseas/antagonistas & inhibidores , Proteínas Cromosómicas no Histona/fisiología , Células Madre Embrionarias/citología , Corazón/embriología , Proteínas Musculares/fisiología , Miocitos Cardíacos/citología , Proteína Nodal/antagonistas & inhibidores , Proteínas/fisiología , Receptores Acoplados a Proteínas G/fisiología , Animales , Receptores de Apelina , Diferenciación Celular , Proliferación Celular , Células Cultivadas , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Citocinas , Ratones , Transducción de Señal , Proteína Smad2/fisiología
9.
Oncotarget ; 4(2): 289-97, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23470527

RESUMEN

The TP73 gene, a member of the p53 family, due to the use of different promoters and alternative splicing, is transcribed into different isoforms with contrasting attributes and which contribute to its functional diversity. Considerable efforts are made to identify the functional diversity of the p73 splicing variants during tumorigenesis. TAp73α and TAp73ß isoforms have been shown to differentially regulate cell cycle progression, differentiation and apoptosis. Interestingly, a particular increase in expression of the TAp73 isoform, in favor of the α splicing variant, has been reported in multiple tumour types. Here, we report a distinctive role for TAp73ß isoform in the control of cell migration and invasion. In fact, TAp73ß- dependent induction of p57(Kip2) expression accounted for inhibitory effects on the actin cytoskeleton dynamics and thereby cancer cell motility. In contrast, TAp73α is not able to induce p57(Kip2) expression, and exhibits a positive effect on actin cytoskeleton dynamics as well as cell migration and invasion. In conclusion, the inhibitory effect on cell migration and invasion of TAp73ß would qualify this distinct p73 isoform as tumor suppressor gene. In contrast, the promoting effect of TAp73α on cell motility and invasion strengthens the potential oncogenic activities of this p73 isoform.


Asunto(s)
Citoesqueleto de Actina/fisiología , Movimiento Celular/fisiología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Proteínas de Unión al ADN/fisiología , Proteínas Nucleares/fisiología , Proteínas Supresoras de Tumor/fisiología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/biosíntesis , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Células HeLa , Humanos , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Isoformas de Proteínas , Activación Transcripcional , Transfección , Proteína Tumoral p73 , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
10.
Placenta ; 34(4): 299-309, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23484914

RESUMEN

The placenta provides critical transport functions between the maternal and fetal circulations during intrauterine development. Formation of this interface is controlled by nuclear transcription factors including homeobox genes. Here we summarize current knowledge regarding the expression and function of homeobox genes in the placenta. We also describe the identification of target transcription factors including PPARγ, biological pathways regulated by homeobox genes and their role in placental development. The role of the nuclear receptor PPARγ, ligands and target genes in human placental development is also discussed. A better understanding of these pathways will improve our knowledge of placental cell biology and has the potential to reveal new molecular targets for the early detection and diagnosis of pregnancy complications including human fetal growth restriction.


Asunto(s)
Genes Homeobox/fisiología , PPAR gamma/genética , Enfermedades Placentarias/patología , Placenta/patología , Placentación , Placentación/genética , Animales , Diferenciación Celular , Células Cultivadas , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Femenino , Retardo del Crecimiento Fetal/genética , Proteínas de Homeodominio/fisiología , Humanos , Ratones , Enfermedades Placentarias/genética , Placentación/fisiología , Embarazo , Proteínas Proto-Oncogénicas c-jun/fisiología , Proteínas Proto-Oncogénicas c-myc/fisiología , Factores de Transcripción/fisiología , Trofoblastos/fisiología
11.
EMBO J ; 31(13): 2952-64, 2012 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-22569127

RESUMEN

The p57(Kip2) cyclin-dependent kinase inhibitor (CDKi) has been implicated in embryogenesis, stem-cell senescence and pathologies, but little is known of its role in cell cycle control. Here, we show that p57(Kip2) is targeted by the p38 stress-activated protein kinase (SAPK). Phosphorylation of p57(Kip2) at T143 by p38 enhances its association with and inhibition of Cdk2, which results in cell-cycle delay upon stress. Genetic inactivation of the SAPK or the CDKi abolishes cell-cycle delay upon osmostress and results in decreased cell viability. Oxidative stress and ionomycin also induce p38-mediated phosphorylation of p57 and cells lacking p38 or p57 display reduced viability to these stresses. Therefore, cell survival to various stresses depends on p57 phosphorylation by p38 that inhibits CDK activity. Together, these findings provide a novel molecular mechanism by which cells can delay cell cycle progression to maximize cell survival upon stress.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Transducción de Señal/fisiología , Animales , Ionóforos de Calcio/farmacología , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Células HEK293 , Humanos , Ionomicina/farmacología , Ratones , Presión Osmótica/efectos de los fármacos , Presión Osmótica/fisiología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Fosforilación/efectos de los fármacos , Fosforilación/fisiología , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
12.
Carcinogenesis ; 32(12): 1897-904, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22002319

RESUMEN

p57 is a multifunctional protein involved in the regulation of tumor formation and development; however, the biological role of p57 in the pathogenesis of hepatocellular carcinoma (HCC) is poorly understood. To explore the role of p57 in the development of HCC, we examined p57 messenger RNA (mRNA) and protein levels in HCC tissues and adjacent non-cancerous tissues by immunohistochemistry, real-time polymerase chain reaction and western blot analysis. Moreover, we generated stable p57 knockdown HCC cell lines to investigate the impact of p57 downregulation on the growth and invasion of HCC in vitro and in vivo. Our results showed that p57 mRNA and protein levels were significantly decreased in human HCC tissues. In addition, this reduction in p57 expression was associated with increased tumor size, more advanced TNM stages, the presence of capsule invasion and extrahepatic metastasis and decreased overall survival time. In human HCC cell lines, p57 downregulation increased the expression of cyclin D1 and CDK2 and enhanced the activities of CDK4/cyclin D1 and CDK2/cyclin E complexes, resulting in increased cellular proliferation and growth of xenografts. Furthermore, p57 downregulation accelerated the invasion of HCC cells in vitro and in vivo by controlling the activity of LIMK1. In conclusion, the downregulation of p57 accelerates the growth and invasion of HCC, indicating that p57 is an important tumor suppressor in HCC. Based on these findings, p57 may be a potential target for HCC prevention and therapy.


Asunto(s)
Carcinoma Hepatocelular/patología , División Celular/fisiología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Regulación hacia Abajo , Neoplasias Hepáticas/patología , Invasividad Neoplásica , Adulto , Anciano , Animales , Línea Celular Tumoral , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Femenino , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos BALB C , Persona de Mediana Edad , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
13.
Mol Cancer Res ; 9(10): 1269-84, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21816904

RESUMEN

p57(Kip2) is a cyclin-dependent kinase inhibitor belonging to the Cip/Kip family, which also includes p21(Cip1) and p27(Kip1). So far, p57(Kip2) is the least-studied Cip/Kip protein, and for a long time its relevance has been related mainly to its unique role in embryogenesis. Moreover, genetic and molecular studies on animal models and patients with Beckwith-Wiedemann syndrome have shown that alterations in CDKN1C (the p57(Kip2) encoding gene) have functional relevance in the pathogenesis of this disease. Recently, a number of investigations have identified and characterized heretofore unexpected roles for p57(Kip2). The protein appears to be critically involved in initial steps of cell and tissue differentiation, and particularly in neuronal development and erythropoiesis. Intriguingly, p27(Kip1), the Cip/Kip member that is most homologous to p57(Kip2), is primarily involved in the process of cell cycle exit. p57(Kip2) also plays a critical role in controlling cytoskeletal organization and cell migration through its interaction with LIMK-1. Furthermore, p57(Kip2) appears to modulate genome expression. Finally, accumulating evidence indicates that p57(Kip2) protein is frequently downregulated in different types of human epithelial and nonepithelial cancers as a consequence of genetic and epigenetic events. In summary, the emerging picture is that several aspects of p57(Kip2)'s functions are only poorly clarified. This review represents an appraisal of the data available on the p57(Kip2) gene and protein structure, and its role in human physiology and pathology. We particularly focus our attention on p57(Kip2) changes in cancers and pharmacological approaches for modulating p57(Kip2) levels.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Neoplasias/metabolismo , Animales , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/metabolismo , Humanos , Ratones , Neoplasias/genética , Neoplasias/patología
14.
Biochim Biophys Acta ; 1816(1): 50-6, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21447370

RESUMEN

Cyclin-dependent kinase inhibitor 1C CDKN1C (p57(KIP2)) regulates several hallmarks of cancer, including apoptosis, cell invasion and metastasis, tumor differentiation and angiogenesis. p57(KIP2) is generally not mutated in cancer, but its expression is downregulated through epigenetic changes such as DNA methylation and repressive histone marks at the promoter. This opens up possibilities for therapeutic intervention through reactivation of p57(KIP2) gene expression. Furthermore, p57(KIP2) has been tested as a prognostic factor for many types of cancer, even differentiating between early and late stage cancer. In this review, the multifunctional tumor suppressor capabilities of p57(KIP2), the mechanisms of p57(KIP2) transcriptional repression in cancer, and the therapeutic potential of reactivation of p57(KIP2) protein expression will be discussed.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Neoplasias/etiología , Animales , Diferenciación Celular , Proliferación Celular , Humanos , Invasividad Neoplásica , Neoplasias/patología , Neoplasias/terapia
15.
Cereb Cortex ; 21(8): 1840-56, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21245411

RESUMEN

Mounting evidence indicates cyclin-dependent kinase (CDK) inhibitors (CKIs) of the Cip/Kip family, including p57(Kip2) and p27(Kip1), control not only cell cycle exit but also corticogenesis. Nevertheless, distinct activities of p57(Kip2) remain poorly defined. Using in vivo and culture approaches, we show p57(Kip2) overexpression at E14.5-15.5 elicits precursor cell cycle exit, promotes transition from proliferation to neuronal differentiation, and enhances process outgrowth, while opposite effects occur in p57(Kip2)-deficient precursors. Studies at later ages indicate p57(Kip2) overexpression also induces precocious glial differentiation, suggesting stage-dependent effects. In embryonic cortex, p57(Kip2) overexpression advances cell radial migration and alters postnatal laminar positioning. While both CKIs induce differentiation, p57(Kip2) was twice as effective as p27(Kip1) in inducing neuronal differentiation and was not permissive to astrogliogenic effects of ciliary neurotrophic factor, suggesting that the CKIs differentially modulate cell fate decisions. At molecular levels, although highly conserved N-terminal regions of both CKIs elicit cycle withdrawal and differentiation, the C-terminal region of p57(Kip2) alone inhibits in vivo migration. Furthermore, p57(Kip2) effects on neurogenesis and gliogenesis require the N-terminal cyclin/CDK binding/inhibitory domains, while previous p27(Kip1) studies report cell cycle-independent functions. These observations suggest p57(Kip2) coordinates multiple stages of corticogenesis and exhibits distinct and common activities compared with related family member p27(Kip1).


Asunto(s)
Diferenciación Celular/fisiología , Inhibición de Migración Celular/fisiología , Corteza Cerebral/embriología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Células-Madre Neurales/enzimología , Neurogénesis/fisiología , Animales , Células Cultivadas , Corteza Cerebral/citología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Femenino , Masculino , Ratones , Ratones Noqueados , Células-Madre Neurales/citología , Embarazo , Ratas , Ratas Sprague-Dawley
16.
Dev Biol ; 350(2): 464-75, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21147088

RESUMEN

Differentiation often requires conversion of analogue signals to a stable binary output through positive feedback. Hedgehog (Hh) signalling promotes myogenesis in the vertebrate somite, in part by raising the activity of muscle regulatory factors (MRFs) of the Myod family above a threshold. Hh is known to enhance MRF expression. Here we show that Hh is also essential at a second step that increases Myod protein activity, permitting it to promote Myogenin expression. Hh acts by inducing expression of cdkn1c (p57(Kip2)) in slow muscle precursor cells, but neither Hh nor Cdkn1c is required for their cell cycle exit. Cdkn1c co-operates with Myod to drive differentiation of several early zebrafish muscle fibre types. Myod in turn up-regulates cdkn1c, thereby providing a positive feedback loop that switches myogenic cells to terminal differentiation.


Asunto(s)
Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Desarrollo de Músculos , Proteína MioD/fisiología , Animales , Diferenciación Celular , Retroalimentación Fisiológica , Proteínas Hedgehog/fisiología , Factor 5 Regulador Miogénico/fisiología , Miogenina/genética , Transducción de Señal , Pez Cebra
17.
Mol Cell Biol ; 30(21): 5057-70, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20805359

RESUMEN

The cortactin oncoprotein is frequently overexpressed in head and neck squamous cell carcinoma (HNSCC), often due to amplification of the encoding gene (CTTN). While cortactin overexpression enhances invasive potential, recent research indicates that it also promotes cell proliferation, but how cortactin regulates the cell cycle machinery is unclear. In this article we report that stable short hairpin RNA-mediated cortactin knockdown in the 11q13-amplified cell line FaDu led to increased expression of the Cip/Kip cyclin-dependent kinase inhibitors (CDKIs) p21(WAF1/Cip1), p27(Kip1), and p57(Kip2) and inhibition of S-phase entry. These effects were associated with increased binding of p21(WAF1/Cip1) and p27(Kip1) to cyclin D1- and E1-containing complexes and decreased retinoblastoma protein phosphorylation. Cortactin regulated expression of p21(WAF1/Cip1) and p27(Kip1) at the transcriptional and posttranscriptional levels, respectively. The direct roles of p21(WAF1/Cip1), p27(Kip1), and p57(Kip2) downstream of cortactin were confirmed by the transient knockdown of each CDKI by specific small interfering RNAs, which led to partial rescue of cell cycle progression. Interestingly, FaDu cells with reduced cortactin levels also exhibited a significant diminution in RhoA expression and activity, together with decreased expression of Skp2, a critical component of the SCF ubiquitin ligase that targets p27(Kip1) and p57(Kip2) for degradation. Transient knockdown of RhoA in FaDu cells decreased expression of Skp2, enhanced the level of Cip/Kip CDKIs, and attenuated S-phase entry. These findings identify a novel mechanism for regulation of proliferation in 11q13-amplified HNSCC cells, in which overexpressed cortactin acts via RhoA to decrease expression of Cip/Kip CDKIs, and highlight Skp2 as a downstream effector for RhoA in this process.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/fisiopatología , Ciclo Celular/genética , Ciclo Celular/fisiología , Cortactina/genética , Cortactina/fisiología , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/genética , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/fisiología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/fisiopatología , Proteína de Unión al GTP rhoA/genética , Proteína de Unión al GTP rhoA/fisiología , Secuencia de Bases , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular , Cromosomas Humanos Par 11/genética , Cortactina/antagonistas & inhibidores , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Cartilla de ADN/genética , Amplificación de Genes , Expresión Génica , Técnicas de Silenciamiento del Gen , Neoplasias de Cabeza y Cuello/patología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , ARN Interferente Pequeño/genética
18.
Front Horm Res ; 38: 15-24, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20616491

RESUMEN

As model of organogenesis, the pituitary gland is a relatively simple tissue; yet, we understand little of the mechanisms that determine organ size, cell number and allocation of cells to different lineages. While the discovery of cell-restricted transcription factors has led to significant insight into the mechanisms controlling differentiation and cell-specific gene expression, we still need to integrate these processes with control of organ development. The identification of pituitary stem cells has suggested mechanisms for maintenance of adult pituitary but these findings again highlight the crucial role of cell cycle control for determination of progenitor and differentiated cell numbers. We recently described the mechanisms for progenitor cell cycle exit in early pituitary development that critically depend on the cell cycle inhibitor p57Kip2. It appears that cell cycle control is independent of differentiation, indicating that separate regulatory mechanisms must be involved in each process. The role of p57Kip2 appears to be restricted to progenitor cell cycle exit and it is rather the related p27Kip1 that prevents re-entry into the cycle of differentiated cells. While these data revealed a new transient intermediate between progenitors and differentiated cells, they also raised new questions and suggested that separate signals may control differentiation and cell cycle.


Asunto(s)
Ciclo Celular , Diferenciación Celular , Hipófisis/embriología , Células Madre/citología , Animales , Inhibidor p27 de las Quinasas Dependientes de la Ciclina , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Proteínas de Homeodominio/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/fisiología , Proteínas de Dominio T Box/fisiología
19.
Dev Biol ; 337(1): 134-46, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19854167

RESUMEN

Sensory hair cells and supporting cells of the mammalian cochlea and vestibular (balance) organs exit the cell cycle during embryogenesis and do not proliferate thereafter. Here, we have studied the mechanisms underlying the maintenance of the postmitotic state and the proliferative capacity of these cells. We provide the first evidence of the role of cyclin D1 in cell cycle regulation in these cells. Cyclin D1 expression disappeared from embryonic hair cells as differentiation started. The expression was transiently upregulated in cochlear hair cells early postnatally, paralleling the spatiotemporal pattern of unscheduled cell cycle re-entry of cochlear hair cells from the p19(Ink4d)/p21(Cip1) compound mutant mice. Cyclin D1 misexpression in vitro in neonatal vestibular HCs from these mutant mice triggered S-phase re-entry. Thus, cyclin D1 suppression is important for hair cell's quiescence, together with the maintained expression of cyclin-dependent kinase inhibitors. In contrast to hair cells, cyclin D1 expression was maintained in supporting cells when differentiation started. The expression continued during the neonatal period when supporting cells have been shown to re-enter the cell cycle upon stimulation with exogenous mitogens. Thereafter, the steep decline in supporting cell's proliferative activity paralleled with cyclin D1 downregulation. Thus, cyclin D1 critically contributes to the proliferative plasticity of supporting cells. These data suggest that targeted cyclin D1 induction in supporting cells might be an avenue for proliferative regeneration in the inner ear.


Asunto(s)
Ciclo Celular , Ciclina D1/fisiología , Proteínas Inhibidoras de las Quinasas Dependientes de la Ciclina/fisiología , Oído Interno/embriología , Células Ciliadas Auditivas/citología , Animales , Proliferación Celular , Cóclea/química , Inhibidor p19 de las Quinasas Dependientes de la Ciclina/análisis , Inhibidor p19 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/análisis , Inhibidor p21 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/análisis , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/análisis , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Antígeno Ki-67/análisis , Ratones , Transducción de Señal , Proteínas Wnt/fisiología , beta Catenina/fisiología
20.
Cell Cycle ; 8(16): 2497-501, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19597343

RESUMEN

The cyclin-dependent kinase inhibitors (CKIs) p27 and p57 are structurally similar, and their biochemical and cellular functions have been thought to be equivalent. However, mice deficient in either p27 or p57 exhibit markedly different phenotypes, suggesting that the in vivo roles of these two proteins might differ. To address this apparent discrepancy, we have generated a knock-in mouse model in which the endogenous p57 gene is replaced by the p27 gene, with p27 thus being expressed instead of p57. This mouse model has provided evidence that p57 functions as a bona fide CKI in vivo and that most of its roles can be performed by p27. Our findings also highlight and provide insight into the question of what determines the distinct cellular responses to abnormal cell cycling induced by the loss of CKIs.


Asunto(s)
Inhibidor p27 de las Quinasas Dependientes de la Ciclina/fisiología , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/fisiología , Animales , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p57 de las Quinasas Dependientes de la Ciclina/genética , Técnicas de Sustitución del Gen , Ratones , Ratones Mutantes , Modelos Biológicos , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...